Conditional Control of Universal CAR T Cells by Cleavable OFF-Switch AdaptorsClick to copy article linkArticle link copied!
- Michael KvorjakMichael KvorjakUPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15232, United StatesDivision of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15232, United StatesDepartment of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United StatesCenter for Systems Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United StatesMore by Michael Kvorjak
- Elisa RuffoElisa RuffoUPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15232, United StatesDivision of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15232, United StatesDepartment of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United StatesCenter for Systems Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United StatesMore by Elisa Ruffo
- Yaniv TivonYaniv TivonDepartment of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United StatesMore by Yaniv Tivon
- Victor SoVictor SoUPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15232, United StatesDivision of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15232, United StatesDepartment of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United StatesCenter for Systems Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United StatesMore by Victor So
- Avani ParikhAvani ParikhUPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15232, United StatesDivision of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15232, United StatesDepartment of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United StatesCenter for Systems Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United StatesMore by Avani Parikh
- Alexander Deiters*Alexander Deiters*Email: [email protected]Center for Systems Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United StatesDepartment of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United StatesMore by Alexander Deiters
- Jason Lohmueller*Jason Lohmueller*Email: [email protected]UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15232, United StatesDivision of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15232, United StatesDepartment of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United StatesCenter for Systems Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United StatesMore by Jason Lohmueller
Abstract
As living drugs, engineered T cell therapies are revolutionizing disease treatment with their unique functional capabilities. However, they suffer from limitations of potentially unpredictable behavior, toxicities, and nontraditional pharmacokinetics. Engineering conditional control mechanisms responsive to tractable stimuli such as small molecules or light is thus highly desirable. We and others previously developed “universal” chimeric antigen receptors (CARs) that interact with coadministered antibody adaptors to direct target cell killing and T cell activation. Universal CARs are of high therapeutic interest due to their ability to simultaneously target multiple antigens on the same disease or different diseases by combining with adaptors to different antigens. Here, we further enhance the programmability and potential safety of universal CAR T cells by engineering OFF-switch adaptors that can conditionally control CAR activity, including T cell activation, target cell lysis, and transgene expression, in response to a small molecule or light stimulus. Moreover, in adaptor combination assays, OFF-switch adaptors were capable of orthogonal conditional targeting of multiple antigens simultaneously, following Boolean logic. OFF-switch adaptors represent a robust new approach for the precision targeting of universal CAR T cells with potential for enhanced safety.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Results
Engineering Conditional Biotin OFF-Switch Adaptors
UV OFF-Switch Adaptor Control of Universal mSA2 CAR T Cells
Small Molecule OFF-Switch Adaptor Control of Universal mSA2 CAR T Cells
OFF-Switch Adaptors Mediate Conditional Control of NFAT-Inducible Transgene Expression
Precision Multiantigen OFF-Switch Control of mSA2 CAR T Cells
Discussion
Methods
Production of Antibody Adaptor Conjugates
mSA2 CAR Expression Vector and Gamma Retrovirus Production
Cell Line Culture
Primary Human T Cell Culture and Retroviral Transduction
Flow Cytometry Staining
mSA2 CAR T Cell Activation Assay
Target Cell Lysis Assay
Combinatorial Target Cell Lysis Assays
Statistical Methods and Data Analysis
Data Availability
All data generated and analyzed during this study are included in this published article and its Supporting Information or are available from the corresponding author upon reasonable request.
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acssynbio.3c00320.
Supplementary figures showing chemical synthesis of the SMcl OFF-switch adaptor; adaptor biotin quantification; mSA2 CAR expression; T cell subset analysis; functional data for rituximab adaptors; cell viability following UV exposure; and adaptor combination lysis control experiments and supplementary methods describing chemical synthesis of the SMcl OFF-switch adaptor and supplementary references (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
This work was supported by NIH grant R01GM142007 (J.L., A.D.). This work benefitted from using the SPECIAL BD LSRFORTESSA funded by NIH 1S10OD011925-01. This project also used the Hillman Cytometry Facility that is supported in part by award P30CA047904. Cartoons in Figures 1, 2A, 3A, 5A, and 6A were created with BioRender.com.
References
This article references 64 other publications.
- 1Gross, G.; Waks, T.; Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl. Acad. Sci. U.S.A. 1989, 86 (24), 10024– 10028, DOI: 10.1073/pnas.86.24.10024Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXntFChsw%253D%253D&md5=3be7a08d7d83af1c25a740bdc0ab670fExpression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificityGross, Gideon; Waks, Tova; Eshhar, ZeligProceedings of the National Academy of Sciences of the United States of America (1989), 86 (24), 10024-8CODEN: PNASA6; ISSN:0027-8424.To design and direct at will the specificity of T cells in a non-major histocompatibility complex (MHC)-restricted manner, chimeric T-cell receptor (TcR) genes composed of the TcR const. (C) domains fused to the antibody's variable (V) domains were constructed and expressed. Genomic expression vectors were constructed contg. the rearranged gene segments coding for the V region domains of the heavy (VH) and light (VL) chains of an anti-2,4,6-trinitrophenyl (TNP) antibody (SP6) spliced to either one of the C-region gene segments of the α or β TcR chains. Following transfection into a cytotoxic T-cell hybridoma, expression of a functional TcR was detected. The chimeric TcR exhibited the idiotope of the Sp6 anti-TNP antibody and endowed the T cells with a non-MHC-restricted response to the hapten TNP. The transfectants specifically killed and produced interleukin 2 in response to TNP-bearing target cells across strain and species barriers. Moreover, such transfectants responded to immobilized TNP-protein conjugates, bypassing the need for cellular processing and presentation. In the particular system employed, both the TNP-binding site and the Sp6 idiotope reside almost exclusively in the VH chain region. Hence, introduction into T cells of TcR genes contg. only the VHSp6 fused to either the Cα or Cβ was sufficient for the expression of a functional surface receptor. Apparently, the VHCα or VHCβ chimeric chains can pair with the endogenous β or α chains of the recipient T cell to form a functional αβ heterodimeric receptor. Thus, this chimeric receptor provides the T cell with an antibody-like specificity and is able to effectively transmit the signal for T-cell activation and execution of its effector function.
- 2Sadelain, M.; Brentjens, R.; Riviere, I. The basic principles of chimeric antigen receptor design. Cancer Discovery 2013, 3 (4), 388– 398, DOI: 10.1158/2159-8290.CD-12-0548Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXlvVCit7c%253D&md5=ebd63df454ec54c889f874c1dc59d669The Basic Principles of Chimeric Antigen Receptor DesignSadelain, Michel; Brentjens, Renier; Riviere, IsabelleCancer Discovery (2013), 3 (4), 388-398CODEN: CDAIB2; ISSN:2159-8274. (American Association for Cancer Research)A review. Chimeric antigen receptors (CAR) are recombinant receptors that provide both antigen-binding and T-cell-activating functions. A multitude of CARs has been reported over the past decade, targeting an array of cell surface tumor antigens. Their biol. functions have dramatically changed following the introduction of tripartite receptors comprising a costimulatory domain, termed second-generation CARs. These have recently shown clin. benefit in patients treated with CD19-targeted autologous T cells. CARs may be combined with costimulatory ligands, chimeric costimulatory receptors, or cytokines to further enhance T-cell potency, specificity, and safety. CARs represent a new class of drugs with exciting potential for cancer immunotherapy. Significance: CARs are a new class of drugs with great potential for cancer immunotherapy. Upon their expression in T lymphocytes, CARs direct potent, targeted immune responses that have recently shown encouraging clin. outcomes in a subset of patients with B-cell malignancies. This review focuses on the design of CARs, including the requirements for optimal antigen recognition and different modalities to provide costimulatory support to targeted T cells, which include the use of second- and third-generation CARs, costimulatory ligands, chimeric costimulatory receptors, and cytokines. Cancer Discov; 3(4); 388-98. ©2013 AACR.
- 3June, C. H.; Sadelain, M. Chimeric Antigen Receptor Therapy. N. Engl. J. Med. 2018, 379 (1), 64– 73, DOI: 10.1056/NEJMra1706169Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlert7rL&md5=87cbf53139fed4142b1529e6f25f0ea5Chimeric antigen receptor therapyJune, Carl H.; Sadelain, MichelNew England Journal of Medicine (2018), 379 (1), 64-73CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)The present article describes about principles of T-cell engineering and synthetic immunity, with focus on efficacy and toxic effects of CAR therapies.
- 4Lim, W. A.; June, C. H. The Principles of Engineering Immune Cells to Treat Cancer. Cell 2017, 168 (4), 724– 740, DOI: 10.1016/j.cell.2017.01.016Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXis1ygtLs%253D&md5=2933e55842cae207c5258ca3934eeb2eThe principles of engineering immune cells to treat cancerLim, Wendell A.; June, Carl H.Cell (Cambridge, MA, United States) (2017), 168 (4), 724-740CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. Chimeric antigen receptor (CAR) T cells have proven that engineered immune cells can serve as a powerful new class of cancer therapeutics. Clin. experience has helped to define the major challenges that must be met to make engineered T cells a reliable, safe, and effective platform that can be deployed against a broad range of tumors. The emergence of synthetic biol. approaches for cellular engineering is providing us with a broadly expanded set of tools for programming immune cells. We discuss how these tools could be used to design the next generation of smart T cell precision therapeutics.
- 5Urbanska, K.; Lanitis, E.; Poussin, M.; Lynn, R. C.; Gavin, B. P.; Kelderman, S.; Yu, J.; Scholler, N.; Powell, D. J. A Universal Strategy for Adoptive Immunotherapy of Cancer through Use of a Novel T-cell Antigen Receptor. Cancer Res. 2012, 72 (7), 1844– 1852, DOI: 10.1158/0008-5472.CAN-11-3890Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XkvVeltrs%253D&md5=b46e16be9bee35426d0a9ff2108bbb6dA Universal Strategy for Adoptive Immunotherapy of Cancer through Use of a Novel T-cell Antigen ReceptorUrbanska, Katarzyna; Lanitis, Evripidis; Poussin, Mathilde; Lynn, Rachel C.; Gavin, Brian P.; Kelderman, Sander; Yu, Jason; Scholler, Nathalie; Powell, Daniel J., Jr.Cancer Research (2012), 72 (7), 1844-1852CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Adoptive immunotherapies composed of T cells engineered to express a chimeric antigen receptor (CAR) offer an attractive strategy for treatment of human cancer. However, CARs have a fixed antigen specificity such that only one tumor-assocd. antigen (TAA) can be targeted, limiting the efficacy that can be achieved because of heterogeneous TAA expression. For this reason, a more generalized and effective application of CAR therapy would benefit from the capability to produce large panels of CARs against many known TAAs. In this study, we show a novel strategy to extend the recognition specificity potential of a bioengineered lymphocyte population, allowing flexible approaches to redirect T cells against various TAAs. Our strategy employs a biotin-binding immune receptor (BBIR) composed of an extracellular-modified avidin linked to an intracellular T-cell signaling domain. BBIR T cells recognized and bound exclusively to cancer cells pretargeted with specific biotinylated mols. The versatility afforded by BBIRs permitted sequential or simultaneous targeting of a combination of distinct antigens. Together, our findings show that a platform of universal T-cell specificity can significantly extend conventional CAR approaches, permitting the tailored generation of T cells of unlimited antigen specificity for improving the effectiveness of adoptive T-cell immunotherapies for cancer. Cancer Res; 72(7); 1844-52.
- 6Lee, D. W.; Kochenderfer, J. N.; Stetler-Stevenson, M.; Cui, Y. K.; Delbrook, C.; Feldman, S. A.; Fry, T. J.; Orentas, R.; Sabatino, M.; Shah, N. N. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 2015, 385 (9967), 517– 528, DOI: 10.1016/S0140-6736(14)61403-3Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhslGjtLbM&md5=6d2ae3cb6d1672a1f0b3c0fedef2c337T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trialLee, Daniel W.; Kochenderfer, James N.; Stetler-Stevenson, Maryalice; Cui, Yongzhi K.; Delbrook, Cindy; Feldman, Steven A.; Fry, Terry J.; Orentas, Rimas; Sabatino, Marianna; Shah, Nirali N.; Steinberg, Seth M.; Stroncek, Dave; Tschernia, Nick; Yuan, Constance; Zhang, Hua; Zhang, Ling; Rosenberg, Steven A.; Wayne, Alan S.; Mackall, Crystal L.Lancet (2015), 385 (9967), 517-528CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)Chimeric antigen receptor (CAR) modified T cells targeting CD19 have shown activity in case series of patients with acute and chronic lymphocytic leukemia and B-cell lymphomas, but feasibility, toxicity, and response rates of consecutively enrolled patients treated with a consistent regimen and assessed on an intention-to-treat basis have not been reported. We aimed to define feasibility, toxicity, max. tolerated dose, response rate, and biol. correlates of response in children and young adults with refractory B-cell malignancies treated with CD19-CAR T cells. This phase 1, dose-escalation trial consecutively enrolled children and young adults (aged 1-30 years) with relapsed or refractory acute lymphoblastic leukemia or non-Hodgkin lymphoma. Autologous T cells were engineered via an 11-day manufg. process to express a CD19-CAR incorporating an anti-CD19 single-chain variable fragment plus TCR zeta and CD28 signalling domains. All patients received fludarabine and cyclophosphamide before a single infusion of CD19-CAR T cells. Using a std. 3 + 3 design to establish the max. tolerated dose, patients received either 1 × 106 CAR-transduced T cells per kg (dose 1), 3 × 106 CAR-transduced T cells per kg (dose 2), or the entire CAR T-cell product if sufficient nos. of cells to meet the assigned dose were not generated. After the dose-escalation phase, an expansion cohort was treated at the max. tolerated dose. The trial is registered with ClinicalTrials.gov, no. NCT01593696. Between July 2, 2012, and June 20, 2014, 21 patients (including eight who had previously undergone allogeneic haematopoietic stem-cell transplantation) were enrolled and infused with CD19-CAR T cells. 19 received the prescribed dose of CD19-CAR T cells, whereas the assigned dose concn. could not be generated for two patients (90% feasible). All patients enrolled were assessed for response. The max. tolerated dose was defined as 1 × 106 CD19-CAR T cells per kg. All toxicities were fully reversible, with the most severe being grade 4 cytokine release syndrome that occurred in three (14%) of 21 patients (95% CI 3·0-36·3). The most common non-haematol. grade 3 adverse events were fever (nine [43%] of 21 patients), hypokalemia (nine [43%] of 21 patients), fever and neutropenia (eight [38%] of 21 patients), and cytokine release syndrome (three [14%) of 21 patients). CD19-CAR T cell therapy is feasible, safe, and mediates potent anti-leukemic activity in children and young adults with chemotherapy-resistant B-precursor acute lymphoblastic leukemia. All toxicities were reversible and prolonged B-cell aplasia did not occur. National Institutes of Health Intramural funds and St Baldrick's Foundation.
- 7Maude, S. L.; Frey, N.; Shaw, P. A.; Aplenc, R.; Barrett, D. M.; Bunin, N. J.; Chew, A.; Gonzalez, V. E.; Zheng, Z.; Lacey, S. F. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014, 371 (16), 1507– 1517, DOI: 10.1056/NEJMoa1407222Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVSls73K&md5=7f13aaa25ae118901d3bec614b07cc3aChimeric antigen receptor T cells for sustained remissions in leukemiaMaude, Shannon L.; Frey, Noelle; Shaw, Pamela A.; Aplenc, Richard; Barrett, David M.; Bunin, Nancy J.; Chew, Anne; Gonzalez, Vanessa E.; Zheng, Zhaohui; Lacey, Simon F.; Mahnke, Yolanda D.; Melenhorst, Jan J.; Rheingold, Susan R.; Shen, Angela; Teachey, David T.; Levine, Bruce L.; June, Carl H.; Porter, David L.; Grupp, Stephan A.New England Journal of Medicine (2014), 371 (16), 1507-1517, 11 pp.CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)Background: Relapsed acute lymphoblastic leukemia (ALL) is difficult to treat despite the availability of aggressive therapies. Chimeric antigen receptor-modified T cells targeting CD19 may overcome many limitations of conventional therapies and induce remission in patients with refractory disease. Methods: We infused autologous T cells transduced with a CD19-directed chimeric antigen receptor (CTL019) lentiviral vector in patients with relapsed or refractory ALL at doses of 0.76 × 106 to 20.6 × 106 CTL019 cells per kg of body wt. Patients were monitored for a response, toxic effects, and the expansion and persistence of circulating CTL019 T cells. Results: A total of 30 children and adults received CTL019. Complete remission was achieved in 27 patients (90%), including 2 patients with blinatumomab-refractory disease and 15 who had undergone stem-cell transplantation. CTL019 cells proliferated in vivo and were detectable in the blood, bone marrow, and cerebrospinal fluid of patients who had a response. Sustained remission was achieved with a 6-mo event-free survival rate of 67% (95% confidence interval [CI], 51 to 88) and an overall survival rate of 78% (95% CI, 65 to 95). At 6 mo, the probability that a patient would have persistence of CTL019 was 68% (95% CI, 50 to 92) and the probability that a patient would have relapse-free B-cell aplasia was 73% (95% CI, 57 to 94). All the patients had the cytokine-release syndrome. Severe cytokine-release syndrome, which developed in 27% of the patients, was assocd. with a higher disease burden before infusion and was effectively treated with the anti-interleukin-6 receptor antibody tocilizumab. Conclusions: Chimeric antigen receptor-modified T-cell therapy against CD19 was effective in treating relapsed and refractory ALL. CTL019 was assocd. with a high remission rate, even among patients for whom stem-cell transplantation had failed, and durable remissions up to 24 mo were obsd.
- 8Park, J. H.; Riviere, I.; Gonen, M.; Wang, X.; Senechal, B.; Curran, K. J.; Sauter, C.; Wang, Y.; Santomasso, B.; Mead, E. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378 (5), 449– 459, DOI: 10.1056/NEJMoa1709919Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlylsbY%253D&md5=a2950f5baba38e254f35f812c56a80e9Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemiaPark, Jae H.; Riviere, Isabelle; Gonen, Mithat; Wang, Xiuyan; Senechal, Brigitte; Curran, Kevin J.; Sauter, Craig; Wang, Yongzeng; Santomasso, Bianca; Mead, Elena; Roshal, Mikhail; Maslak, Peter; Davila, Marco; Brentjens, Renier J.; Sadelain, MichelNew England Journal of Medicine (2018), 378 (5), 449-459CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)CD19-specific chimeric antigen receptor (CAR) T cells induce high rates of initial response among patients with relapsed B-cell acute lymphoblastic leukemia (ALL) and long-term remissions in a subgroup of patients. We conducted a phase 1 trial involving adults with relapsed B-cell ALL who received an infusion of autologous T cells expressing the 19-28z CAR at the Memorial Sloan Kettering Cancer Center (MSKCC). Safety and long-term outcomes were assessed, as were their assocns. with demog., clin., and disease characteristics. A total of 53 adults received 19-28z CAR T cells that were manufd. at MSKCC. After infusion, severe cytokine release syndrome occurred in 14 of 53 patients (26%; 95% confidence interval [CI], 15 to 40); 1 patient died. Complete remission was obsd. in 83% of the patients. At a median follow-up of 29 mo (range, 1 to 65), the median event-free survival was 6.1 mo (95% CI, 5.0 to 11.5), and the median overall survival was 12.9 mo (95% CI, 8.7 to 23.4). Patients with a low disease burden (<5% bone marrow blasts) before treatment had markedly enhanced remission duration and survival, with a median event-free survival of 10.6 mo (95% CI, 5.9 to not reached) and a median overall survival of 20.1 mo (95% CI, 8.7 to not reached). Patients with a higher burden of disease (≥5% bone marrow blasts or extramedullary disease) had a greater incidence of the cytokine release syndrome and neurotoxic events and shorter long-term survival than did patients with a low disease burden. In the entire cohort, the median overall survival was 12.9 mo. Among patients with a low disease burden, the median overall survival was 20.1 mo and was accompanied by a markedly lower incidence of the cytokine release syndrome and neurotoxic events after 19-28z CAR T-cell infusion than was obsd. among patients with a higher disease burden.
- 9Munshi, N. C.; Anderson, L. D., Jr; Shah, N.; Madduri, D.; Berdeja, J.; Lonial, S.; Raje, N.; Lin, Y.; Siegel, D.; Oriol, A. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N. Engl. J. Med. 2021, 384 (8), 705– 716, DOI: 10.1056/NEJMoa2024850Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXlt1KgsLY%253D&md5=730eaa169adca5221b86ece1884c18f3Idecabtagene vicleucel in relapsed and refractory multiple myelomaMunshi, Nikhil C.; Anderson, Larry D., Jr.; Shah, Nina; Madduri, Deepu; Berdeja, Jesus; Lonial, Sagar; Raje, Noopur; Lin, Yi; Siegel, David; Oriol, Albert; Moreau, Philippe; Yakoub-Agha, Ibrahim; Delforge, Michel; Cavo, Michele; Einsele, Hermann; Goldschmidt, Hartmut; Weisel, Katja; Rambaldi, Alessandro; Reece, Donna; Petrocca, Fabio; Massaro, Monica; Connarn, Jamie N.; Kaiser, Shari; Patel, Payal; Huang, Liping; Campbell, Timothy B.; Hege, Kristen; San-Miguel, JesusNew England Journal of Medicine (2021), 384 (8), 705-716CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)Idecabtagene vicleucel (ide-cel, also called bb2121), a B-cell maturation antigen-directed chimeric antigen receptor (CAR) T-cell therapy, has shown clin. activity with expected CAR T-cell toxic effects in patients with relapsed and refractory multiple myeloma. methods In this phase 2 study, we sought to confirm the efficacy and safety of ide-cel in patients with relapsed and refractory myeloma. Patients with disease after at least three previous regimens including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 antibody were enrolled. Patients received ide-cel target doses of 150 x 106 to 450 x 106 CAR-pos. (CAR+) T cells. The primary end point was an overall response (partial response or better); a key secondary end point was a complete response or better (comprising complete and stringent complete responses). results Of 140 patients enrolled, 128 received ide-cel. At a median follow-up of 13.3 mo, 94 of 128 patients (73%) had a response, and 42 of 128 (33%) had a complete response or better. Minimal residual disease (MRD)-neg. status (<10-5 nucleated cells) was confirmed in 33 patients, representing 26% of all 128 patients who were treated and 79% of the 42 patients who had a complete response or better. The median progression-free survival was 8.8 mo (95% confidence interval, 5.6 to 11.6). Common toxic effects among the 128 treated patients included neutropenia in 117 patients (91%), anemia in 89 (70%), and thrombocytopenia in 81 (63%). Cytokine release syndrome was reported in 107 patients (84%), including 7 (5%) who had events of grade 3 or higher. Neurotoxic effects developed in 23 patients (18%) and were of grade 3 in 4 patients (3%); no neurotoxic effects higher than grade 3 occurred. Cellular kinetic anal. confirmed CAR+ T cells in 29 of 49 patients (59%) at 6 mo and 4 of 11 patients (36%) at 12 mo after infusion. conclusions Ide-cel induced responses in a majority of heavily pretreated patients with refractory and relapsed myeloma; MRD-neg. status was achieved in 26% of treated patients. Almost all patients had grade 3 or 4 toxic effects, most commonly hematol. toxic effects and cytokine release syndrome.
From NLM Medline
- 10Ramakrishna, S.; Barsan, V.; Mackall, C. Prospects and challenges for use of CAR T cell therapies in solid tumors. Expert Opin. Biol. Ther. 2020, 20 (5), 503– 516, DOI: 10.1080/14712598.2020.1738378Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvVertLo%253D&md5=d9328ebe64880ed45af37bd1b84aa7cbProspects and challenges for use of CAR T cell therapies in solid tumorsRamakrishna, Sneha; Barsan, Valentin; Mackall, CrystalExpert Opinion on Biological Therapy (2020), 20 (5), 503-516CODEN: EOBTA2; ISSN:1471-2598. (Taylor & Francis Ltd.)A review. : Chimeric antigen receptor (CAR) T cell therapy has provided patients with relapsed/refractory B cell malignancies a new therapeutic option, but this class of therapeutics has not demonstrated consistent therapeutic benefit in solid tumors.: Here we review the literature to identify numerous factors that contribute to this discrepancy, using pediatric cancers as a platform to understand these limitations. We discuss an inability to target highly and homogenously expressed lineage-assocd. antigens due to risks of on-target off-tumor toxicity, T cell dysfunction related to T cell exhaustion and the suppressive tumor microenvironment (TME), and inefficient CAR T cell trafficking into solid tumors. As our understanding of the biol. of CAR T cells improves and innovations in engineering CAR platforms emerge, next generation CAR T cell therapeutics designed to overcome these challenges will enter the clinic for testing.: New approaches to address the challenges that have limited the efficacy of CAR T cell therapeutics in solid tumors are emerging. These approaches include next-generation CAR T cell engineering to overcome antigen heterogeneity, to mitigate T cell exhaustion and to prevent suppression by the TME, as well as novel approaches for regional delivery to facilitate tumor T cell trafficking.
- 11Maldini, C. R.; Ellis, G. I.; Riley, J. L. CAR T cells for infection, autoimmunity and allotransplantation. Nat. Rev. Immunol. 2018, 18 (10), 605– 616, DOI: 10.1038/s41577-018-0042-2Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlyrurbF&md5=ab83fb3b4a2abac6b439fbc5e560ac90CAR T cells for infection, autoimmunity and allotransplantationMaldini, Colby R.; Ellis, Gavin I.; Riley, James L.Nature Reviews Immunology (2018), 18 (10), 605-616CODEN: NRIABX; ISSN:1474-1733. (Nature Research)Chimeric antigen receptors (CARs) have shown remarkable ability to re-direct T cells to target CD19-expressing tumors, resulting in remission rates of up to 90% in individuals with paediatric acute lymphoblastic lymphoma. Lessons learned from these clin. trials of adoptive T cell therapy for cancer, as well as investments made in manufg. T cells at com. scale, have inspired researchers to develop CARs for addnl. applications. Here, we explore the challenges and opportunities of using this technol. to target infectious diseases such as with HIV and undesired immune responses such as autoimmunity and transplant rejection. Despite substantial obstacles, the potential of CAR T cells to enable cures for a wide array of disease settings could be transformational for the medical field.
- 12Bertoletti, A.; Tan, A. T. Challenges of CAR- and TCR-T cell-based therapy for chronic infections. J. Exp. Med. 2020, 217 (5), e20191663 DOI: 10.1084/jem.20191663Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXovFSntrs%253D&md5=86d61cc45857a869f78b0525abefe18dChallenges of CAR- and TCR-T cell-based therapy for chronic infectionsBertoletti, Antonio; Tan, Anthony TanotoJournal of Experimental Medicine (2020), 217 (5), e20191663CODEN: JEMEAV; ISSN:1540-9538. (Rockefeller University Press)A review. While therapy with T cells engineered with a chimeric antigen receptor (CAR) or a classical T cell receptor (TCR) is revolutionizing cancer treatment, its adoption in infectious diseases has been met with considerable resistance. Can we find its value for the cure of infections.
- 13Press, M. F.; Cordon-Cardo, C.; Slamon, D. J. Expression of the HER-2/neu proto-oncogene in normal human adult and fetal tissues. Oncogene 1990, 5 (7), 953– 962Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXkvFalsbc%253D&md5=1c839969b674279e8820f7575c2ecae7Expression of the HER-2/neu proto-oncogene in normal human adult and fetal tissuesPress, Michael F.; Cordon-Cardo, Carlos; Slamon, Dennis J.Oncogene (1990), 5 (7), 953-62CODEN: ONCNES; ISSN:0950-9232.The HER-2/neu proto-oncogene is homologous with, but distinct from the epidermal growth factor receptor. A variety of normal adult and fetal tissues were evaluated for HER-2/neu expression using immunohistochem. and Northern blot analyses to identify the HER-2/neu protein and transcript resp.. HER-2/neu protein was identified on cell membranes of epithelial cells in the gastrointestinal, respiratory, reproductive, and urinary tract as well as in the skin, breast and placenta. Northern hybridization confirmed the presence of the 4.5 kb transcript encoding the protein in these tissues. The amt. of HER-2/neu message and protein was generally higher in fetal tissues than in the corresponding normal adult tissues. HER-2/neu expression levels in these normal tissues were similar to the levels found in non-amplified, non-overexpresing breast cancers and breast cancer cell lines. Southern hybridization of extd. DNA showed that none of the normal tissues expressing HER-2/neu had amplification of the gene. These results confirm that HER-2/neu is normally a membrane constituent of a variety of epithelial cell types.
- 14Yano, S.; Kondo, K.; Yamaguchi, M.; Richmond, G.; Hutchison, M.; Wakeling, A.; Averbuch, S.; Wadsworth, P. Distribution and function of EGFR in human tissue and the effect of EGFR tyrosine kinase inhibition. Anticancer Res. 2003, 23 (5A), 3639– 3650Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXkvFWn&md5=70a289e6c8c5492e9c3537d77880af8dDistribution and function of EGFR in human tissue and the effect of EGFR tyrosine kinase inhibitionYano, Seiichi; Kondo, Kaoru; Yamaguchi, Motonori; Richmond, Graham; Hutchison, Michael; Wakeling, Alan; Averbuch, Steven; Wadsworth, PeterAnticancer Research (2003), 23 (5A), 3639-3650CODEN: ANTRD4; ISSN:0250-7005. (International Institute of Anticancer Research)A review. From immunohistochem. and ligand-binding studies, it is known that the epidermal growth factor receptor (EGFR), a member of the erbB family of receptors, is expressed in tissues of epithelial, mesenchymal and neuronal origin and plays a major role in normal cellular processes such as proliferation, differentiation and development. EGFR is highly expressed in a no. of solid tumors and its expression correlates with tumor progression, resistance to chemotherapy and a poor prognosis; it is consequently an attractive target for the rational design of novel anticancer agents. Knowledge of the role of EGFR in normal tissues will help the understanding of the adverse events assocd. with such agents. Studies in knockout mice and preclin. toxicol. studies have shown that the major effects of inhibiting the EGFR are skin and, gastrointestinal toxicities. Clin. studies with inhibitors of EGFR, such as gefitinib, cetuximab and erlotinib, have shown a favorable adverse-event profile, primarily consisting of skin and gastrointestinal toxicities, as predicted from the mechanism-based effects obsd. in preclin. studies.
- 15Majzner, R. G.; Mackall, C. L. Clinical lessons learned from the first leg of the CAR T cell journey. Nat. Med. 2019, 25 (9), 1341– 1355, DOI: 10.1038/s41591-019-0564-6Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslelurbN&md5=ab243f9a176dc0e3f9d65aebb9097021Clinical lessons learned from the first leg of the CAR T cell journeyMajzner, Robbie G.; Mackall, Crystal L.Nature Medicine (New York, NY, United States) (2019), 25 (9), 1341-1355CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)Chimeric antigen receptor (CAR) T cell therapy for B cell malignancies has surpassed expectations, driving an ever-expanding no. of clin. trials and the first US Food and Drug Administration approvals of cell therapies for the treatment of cancer. This experience has illuminated some generalizable requirements for CAR T cell efficacy as well as the interplay between disease biol. and clin. outcomes. Major CAR intrinsic variables affecting T cell behavior have been defined, and mechanisms of tumor resistance are increasingly understood. Here, we review the clin. experience with CAR T cells amassed to date, including but not limited to B cell malignancies, emphasizing factors assocd. with efficacy, resistance and major barriers to success. We also discuss how these insights are driving next-generation clin. trials, including those in solid tumors.
- 16Kershaw, M. H.; Westwood, J. A.; Parker, L. L.; Wang, G.; Eshhar, Z.; Mavroukakis, S. A.; White, D. E.; Wunderlich, J. R.; Canevari, S.; Rogers-Freezer, L. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin. Cancer Res. 2006, 12 (20), 6106– 6115, DOI: 10.1158/1078-0432.CCR-06-1183Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFWhsbnL&md5=a6a439a0187f50d8a22e12dfc1da89e5A Phase I Study on Adoptive Immunotherapy Using Gene-Modified T Cells for Ovarian CancerKershaw, Michael H.; Westwood, Jennifer A.; Parker, Linda L.; Wang, Gang; Eshhar, Zelig; Mavroukakis, Sharon A.; White, Donald E.; Wunderlich, John R.; Canevari, Silvana; Rogers-Freezer, Linda; Chen, Clara C.; Yang, James C.; Rosenberg, Steven A.; Hwu, PatrickClinical Cancer Research (2006), 12 (20, Pt. 1), 6106-6115CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: A phase I study was conducted to assess the safety of adoptive immunotherapy using gene-modified autologous T cells for the treatment of metastatic ovarian cancer. Exptl. Design: T cells with reactivity against the ovarian cancer-assocd. antigen α-folate receptor (FR) were generated by genetic modification of autologous T cells with a chimeric gene incorporating an anti-FR single-chain antibody linked to the signaling domain of the Fc receptor γ chain. Patients were assigned to one of two cohorts in the study. Eight patients in cohort 1 received a dose escalation of T cells in combination with high-dose interleukin-2, and six patients in cohort 2 received dual-specific T cells (reactive with both FR and allogeneic cells) followed by immunization with allogeneic peripheral blood mononuclear cells. Results: Five patients in cohort 1 experienced some grade 3 to 4 treatment-related toxicity that was probably due to interleukin-2 administration, which could be managed using std. measures. Patients in cohort 2 experienced relatively mild side effects with grade 1 to 2 symptoms. No redn. in tumor burden was seen in any patient. Tracking 111In-labeled adoptively transferred T cells in cohort 1 revealed a lack of specific localization of T cells to tumor except in one patient where some signal was detected in a peritoneal deposit. PCR anal. showed that gene-modified T cells were present in the circulation in large nos. for the first 2 days after transfer, but these quickly declined to be barely detectable 1 mo later in most patients. An inhibitory factor developed in the serum of three of six patients tested over the period of treatment, which significantly reduced the ability of gene-modified T cells to respond against FR+ tumor cells. Conclusions: Large nos. of gene-modified tumor-reactive T cells can be safely given to patients, but these cells do not persist in large nos. long term. Future studies need to employ strategies to extend T cell persistence. This report is the first to document the use of genetically redirected T cells for the treatment of ovarian cancer.
- 17Lamers, C. H.; Sleijfer, S.; Vulto, A. G.; Kruit, W. H.; Kliffen, M.; Debets, R.; Gratama, J. W.; Stoter, G.; Oosterwijk, E. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J. Clin. Oncol. 2006, 24 (13), e20– e22, DOI: 10.1200/JCO.2006.05.9964Google ScholarThere is no corresponding record for this reference.
- 18Maus, M. V.; Haas, A. R.; Beatty, G. L.; Albelda, S. M.; Levine, B. L.; Liu, X.; Zhao, Y.; Kalos, M.; June, C. H. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol. Res. 2013, 1 (1), 26– 31, DOI: 10.1158/2326-6066.CIR-13-0006Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtFSjtLo%253D&md5=39a36c1c1c297760ad7f7298c09df899T cells expressing chimeric antigen receptors can cause anaphylaxis in humansMaus, Marcela V.; Haas, Andrew R.; Beatty, Gregory L.; Albelda, Steven M.; Levine, Bruce L.; Liu, Xiaojun; Zhao, Yangbing; Kalos, Michael; June, Carl H.Cancer Immunology Research (2013), 1 (1), 26-31CODEN: CIRACV; ISSN:2326-6066. (American Association for Cancer Research)T cells can be redirected to overcome tolerance to cancer by engineering with integrating vectors to express a chimeric antigen receptor (CAR). In preclin. models, we have previously shown that transfection of T cells with mRNA coding for a CAR is an alternative strategy that has antitumor efficacy and the potential to evaluate the on-target off-tumor toxicity of new CAR targets safely due to transient mRNA CAR expression. Here, we report the safety obsd. in four patients treated with autologous T cells that had been electroporated with mRNA coding for a CAR derived from a murine antibody to human mesothelin. Because of the transient nature of CAR expression on the T cells, subjects in the clin. study were given repeated infusions of the CAR-T cells to assess their safety. One subject developed anaphylaxis and cardiac arrest within minutes of completing the third infusion. Although human anti-mouse Ig (Ig)G antibodies have been known to develop with CAR-transduced T cells, they have been thought to have no adverse clin. consequences. This is the first description of clin. anaphylaxis resulting from CAR-modified T cells, most likely through IgE antibodies specific to the CAR. These results indicate that the potential immunogenicity of CARs derived from murine antibodies may be a safety issue for mRNA CARs, esp. when administered using an intermittent dosing schedule.
- 19Roybal, K. T.; Rupp, L. J.; Morsut, L.; Walker, W. J.; McNally, K. A.; Park, J. S.; Lim, W. A. Precision Tumor Recognition by T Cells With Combinatorial Antigen-Sensing Circuits. Cell 2016, 164 (4), 770– 779, DOI: 10.1016/j.cell.2016.01.011Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xhs12gt78%253D&md5=dc3da3e426700a1dd4f57417155c97b5Precision Tumor Recognition by T Cells With Combinatorial Antigen-Sensing CircuitsRoybal, Kole T.; Rupp, Levi J.; Morsut, Leonardo; Walker, Whitney J.; McNally, Krista A.; Park, Jason S.; Lim, Wendell A.Cell (Cambridge, MA, United States) (2016), 164 (4), 770-779CODEN: CELLB5; ISSN:0092-8674. (Cell Press)T cells can be re-directed to kill cancer cells using chimeric antigen receptors (CARs) or T cell receptors (TCRs). This approach, however, is constrained by the rarity of tumor-specific single antigens. Targeting antigens also found on bystander tissues can cause life-threatening adverse effects. A powerful way to enhance ON-target activity of therapeutic T cells is to engineer them to require combinatorial antigens. Here, the authors engineer a combinatorially activated T cell circuit in which a synthetic Notch receptor for one antigen induces the expression of a CAR for a second antigen. These dual-receptor AND-gate T cells are only armed and activated in the presence of dual antigen tumor cells. These T cells show precise therapeutic discrimination in vivo-sparing single antigen "bystander" tumors while efficiently clearing combinatorial antigen "disease" tumors. This type of precision dual-receptor circuit opens the door to immune recognition of a wider range of tumors.
- 20Esensten, J. H.; Bluestone, J. A.; Lim, W. A. Engineering Therapeutic T Cells: From Synthetic Biology to Clinical Trials. Annu. Rev. Pathol 2017, 12, 305– 330, DOI: 10.1146/annurev-pathol-052016-100304Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVyis7vN&md5=601808024db94dc019410b9b0026bdeaEngineering Therapeutic T Cells: From Synthetic Biology to Clinical TrialsEsensten, Jonathan H.; Bluestone, Jeffrey A.; Lim, Wendell A.Annual Review of Pathology: Mechanisms of Disease (2017), 12 (), 305-330CODEN: ARPMCU; ISSN:1553-4006. (Annual Reviews)Engineered T cells are currently in clin. trials to treat patients with cancer, solid organ transplants, and autoimmune diseases. However, the field is still in its infancy. The design, and manufg., of T cell therapies is not standardized and is performed mostly in academic settings by competing groups. Reliable methods to define dose and pharmacokinetics of T cell therapies need to be developed. As of mid-2016, there are no US Food and Drug Administration (FDA)-approved T cell therapeutics on the market, and FDA regulations are only slowly adapting to the new technologies. Further development of engineered T cell therapies requires advances in immunol., synthetic biol., manufg. processes, and government regulation. In this review, we outline some of these challenges and discuss the contributions that pathologists can make to this emerging field.
- 21Cho, J. H.; Collins, J. J.; Wong, W. W. Universal Chimeric Antigen Receptors for Multiplexed and Logical Control of T Cell Responses. Cell 2018, 173 (6), 1426– 1438.e11, DOI: 10.1016/j.cell.2018.03.038Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXosFWhsLs%253D&md5=4e6791c20148e208a9eb9ac031d351eaUniversal Chimeric Antigen Receptors for Multiplexed and Logical Control of T Cell ResponsesCho, Jang Hwan; Collins, James J.; Wong, Wilson W.Cell (Cambridge, MA, United States) (2018), 173 (6), 1426-1438.e11CODEN: CELLB5; ISSN:0092-8674. (Cell Press)T cells expressing chimeric antigen receptors (CARs) are promising cancer therapeutic agents, with the prospect of becoming the ultimate smart cancer therapeutics. To expand the capability of CAR T cells, here, we present a split, universal, and programmable (SUPRA) CAR system that simultaneously encompasses multiple crit. "upgrades," such as the ability to switch targets without re-engineering the T cells, finely tune T cell activation strength, and sense and logically respond to multiple antigens. These features are useful to combat relapse, mitigate over-activation, and enhance specificity. We test our SUPRA system against two different tumor models to demonstrate its broad utility and humanize its components to minimize potential immunogenicity concerns. Furthermore, we extend the orthogonal SUPRA CAR system to regulate different T cell subsets independently, demonstrating a dually inducible CAR system. Together, these SUPRA CARs illustrate that multiple advanced logic and control features can be implemented into a single, integrated system.
- 22Kudo, K.; Imai, C.; Lorenzini, P.; Kamiya, T.; Kono, K.; Davidoff, A.; Chng, W.; Campana, D. T lymphocytes expressing a CD16 signaling receptor exert antibody-dependent cancer cell killing. Cancer Res. 2014, 74 (1), 93– 103, DOI: 10.1158/0008-5472.CAN-13-1365Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXislKhtQ%253D%253D&md5=997922da6b258eb409360d48e8bdcbb9T Lymphocytes Expressing a CD16 Signaling Receptor Exert Antibody-Dependent Cancer Cell KillingKudo, Ko; Imai, Chihaya; Lorenzini, Paolo; Kamiya, Takahiro; Kono, Koji; Davidoff, Andrew M.; Chng, Wee Joo; Campana, DarioCancer Research (2014), 74 (1), 93-103CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)To expand applications for T-cell-based immunotherapy in cancer, we designed a receptor that binds the Fc portion of human Igs and delivers activation signals. The construct included the high-affinity CD16 (FCGR3A) V158 variant, CD8α hinge, and transmembrane domains, along with signaling domains from CD3ζ and 4-1BB (TNFRSF9), forming a chimeric receptor termed CD16V-BB-ζ After retrovirus-mediated expression in human T cells, CD16V-BB-ζ- bound humanized antibodies with higher affinity than a control receptor contg. the more common F158 variant. Engagement of CD16V-VV-ζ provoked T-cell activation, exocytosis of lytic granules, and sustained proliferation, with a mean cell recovery after 4-wk coculture with Daudi lymphoma cells and rituximab of nearly 70-fold relative to input cells. In contrast, unbound antibody alone produced no effect. CD16-BB-ζ T cells specifically killed lymphoma cells and primary chronic lymphocytic leukemia cells in combination with rituximab at a low effectontarget ratio, even when assayed on mesenchymal cells. Trastuzumab triggered CD16V-BB-ζ-mediated killing of HER2 (ERBB2)+ breast and gastric cancer cells; similar results were obtained with an anti-GD2 antibody in neuroblastoma and osteosarcoma cells. Furthermore, coadministration of CD16V-BB-ζ T cells with immunotherapeutic antibodies exerted considerable antitumor activity in vivo. Signaling mediated by 4-1BB-CD3ζ induced higher T-cell activation, proliferation, and cytotoxicity than 3 or FcεRIγ, and the receptor was expressed effectively after mRNA electroporation without viral vectors, facilitating clin. translation. Our results offer preclin. proof of concept for CD16V-BB-ζ as a universal, next-generation chimeric receptor with the potential to augment the efficacy of antibody therapies for cancer.
- 23Minutolo, N. G.; Hollander, E. E.; Powell, D. J., Jr The Emergence of Universal Immune Receptor T Cell Therapy for Cancer. Front. Oncol. 2019, 9, 176, DOI: 10.3389/fonc.2019.00176Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M%252Fltl2qsA%253D%253D&md5=05413c83ff8ab6c6fddacd1b16d7c2d9The Emergence of Universal Immune Receptor T Cell Therapy for CancerMinutolo Nicholas G; Hollander Erin E; Powell Daniel J Jr; Minutolo Nicholas G; Minutolo Nicholas G; Minutolo Nicholas G; Hollander Erin E; Powell Daniel J Jr; Hollander Erin EFrontiers in oncology (2019), 9 (), 176 ISSN:2234-943X.Chimeric antigen receptor (CAR) T cells have shown great success in the treatment of CD19+ hematological malignancies, leading to their recent approval by the FDA as a new cancer treatment modality. However, their broad use is limited since a CAR targets a single tumor associated antigen (TAA), which is not effective against tumors with heterogeneous TAA expression or emerging antigen loss variants. Further, stably engineered CAR T cells can continually and uncontrollably proliferate and activate in response to antigen, potentially causing fatal on-target off-tumor toxicity, cytokine release syndrome, or neurotoxicity without a method of control or elimination. To address these issues, our lab and others have developed various universal immune receptors (UIRs) that allow for targeting of multiple TAAs by T cells expressing a single receptor. UIRs function through the binding of an extracellular adapter domain which acts as a bridge between intracellular T cell signaling domains and a soluble tumor antigen targeting ligand (TL). The dissociation of TAA targeting and T cell signaling confers many advantages over standard CAR therapy, such as dose control of T cell effector function, the ability to simultaneously or sequentially target multiple TAAs, and control of immunologic synapse geometry. There are currently four unique UIR platform types: ADCC-mediating Fc-binding immune receptors, bispecific protein engaging immune receptors, natural binding partner immune receptors, and anti-tag CARs. These UIRs all allow for potential benefits over standard CARs, but also bring unique engineering challenges that will have to be addressed to achieve maximal efficacy and safety in the clinic. Still, UIRs present an exciting new avenue for adoptive T cell transfer therapies and could lead to their expanded use in areas which current CAR therapies have failed. Here we review the development of each UIR platform and their unique functional benefits, and detail the potential hurdles that may need to be overcome for continued clinical translation.
- 24Ruffo, E.; Butchy, A. A.; Tivon, Y.; So, V.; Kvorjak, M.; Parikh, A.; Adams, E. L.; Miskov-Zivanov, N.; Finn, O. J.; Deiters, A. Post-translational covalent assembly of CAR and synNotch receptors for programmable antigen targeting. Nat. Commun. 2023, 14 (1), 2463, DOI: 10.1038/s41467-023-37863-5Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXpsl2iurY%253D&md5=5f0895c2abd463a1ea1ce95ae02196a5Post-translational covalent assembly of CAR and synNotch receptors for programmable antigen targetingRuffo, Elisa; Butchy, Adam A.; Tivon, Yaniv; So, Victor; Kvorjak, Michael; Parikh, Avani; Adams, Eric L.; Miskov-Zivanov, Natasa; Finn, Olivera J.; Deiters, Alexander; Lohmueller, JasonNature Communications (2023), 14 (1), 2463CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)Chimeric antigen receptors (CARs) and synthetic Notch (synNotch) receptors are engineered cell-surface receptors that sense a target antigen and respond by activating T cell receptor signaling or a customized gene program, resp. Here, to expand the targeting capabilities of these receptors, we develop "universal" receptor systems for which receptor specificity can be directed post-translationally via covalent attachment of a co-administered antibody bearing a benzylguanine (BG) motif. A SNAPtag self-labeling enzyme is genetically fused to the receptor and reacts with BG-conjugated antibodies for covalent assembly, programming antigen recognition. We demonstrate that activation of SNAP-CAR and SNAP-synNotch receptors can be successfully targeted by clin. relevant BG-conjugated antibodies, including anti-tumor activity of SNAP-CAR T cells in vivo in a human tumor xenograft mouse model. Finally, we develop a math. model to better define the parameters affecting universal receptor signaling. SNAP receptors provide a powerful strategy to post-translationally reprogram the targeting specificity of engineered cells.
From NLM Medline
- 25Lohmueller, J. J.; Ham, J. D.; Kvorjak, M.; Finn, O. J. mSA2 affinity-enhanced biotin-binding CAR T cells for universal tumor targeting. Oncoimmunology 2018, 7 (1), e1368604 DOI: 10.1080/2162402X.2017.1368604Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhslaltL%252FJ&md5=011672c41c0ebb99f473cd3328f9dfc3mSA2 affinity-enhanced biotin-binding CAR T cells for universal tumor targetingLohmueller, Jason J.; Ham, James D.; Kvorjak, Michael; Finn, Olivera J.OncoImmunology (2018), 7 (1), e1368604/1-e1368604/6CODEN: ONCOGX; ISSN:2162-402X. (Taylor & Francis, Inc.)Chimeric antigen receptor T cells (CAR-Ts) are promising cancer therapeutics. However, since cancer cells can lose the CAR-targeted antigen and avoid destruction, targeting multiple antigens with multiple CARs has been proposed. We illustrate here a less cumbersome alternative, anti-tag CARs (AT-CARs) that bind to tags on tumor-targeting antibodies. We have created novel AT-CARs, using the affinity-enhanced monomeric streptavidin 2 (mSA2) biotin-binding domain that when expressed on T cells can target cancer cells coated with biotinylated antibodies. Human T cells expressing mSA2 CARs with CD28-CD3ζ and 4-1BB-CD3ζ signaling domains were activated by plate-immobilized biotin and by tumor cells coated with biotinylated antibodies against the tumor-assocd. antigens CD19 and CD20. Furthermore, mSA2 CAR T cells were capable of mediating cancer cell lysis and IFNγ prodn. in an antibody dose-dependent manner. The mSA2 CAR is a universal AT-CAR that can be combined with biotinylated tumor-specific antibodies to potentially target many different tumor types.
- 26Tamada, K.; Geng, D.; Sakoda, Y.; Bansal, N.; Srivastava, R.; Li, Z.; Davila, E. Redirecting gene-modified T cells toward various cancer types using tagged antibodies. Clin. Cancer Res. 2012, 18 (23), 6436– 6445, DOI: 10.1158/1078-0432.CCR-12-1449Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhslKgurjJ&md5=93e3532e765309253195922cc35ea078Redirecting Gene-Modified T Cells toward Various Cancer Types Using Tagged AntibodiesTamada, Koji; Geng, Degui; Sakoda, Yukimi; Bansal, Navneeta; Srivastava, Ratika; Li, Zhaoyang; Davila, EduardoClinical Cancer Research (2012), 18 (23), 6436-6445CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: To develop an adaptable gene-based vector that will confer immune cell specificity to various cancer types. Exptl. Design: Human and mouse T cells were genetically engineered to express a chimeric antigen receptor (CAR) that binds a fluorescein isothiocyanate (FITC) mol., termed anti-FITC CAR T cells. Various antibodies (Ab) currently in clin. use including cetuximab (Ctx), trastuzumab (Her2), and rituximab (Rtx) were conjugated with FITC and tested for their ability to bind tumor cells, activate T cells, and induce antitumor effects in vitro and in vivo. Results: Anti-FITC CAR T cells recognize various cancer types when bound with FITC-labeled Abs resulting in efficient target lysis, T-cell proliferation, and cytokine/chemokine prodn. The treatment of immunocompromised mice with human anti-FITC CAR T cells plus FITC-labeled cetuximab (FITC-Ctx) delayed the growth of colon cancer but unexpectedly led to the outgrowth of EGF receptor (EGFR)-neg. tumor cells. On the other hand, in a human pancreatic cancer cell line with uniform EGFR expression, anti-FITC CAR T cells plus FITC-Ctx eradicated preestablished late-stage tumors. In immunocompetent mice, anti-FITC CAR T cells exhibited potent antitumor activity against syngeneic mouse breast cancer expressing Her2 and B-cell lymphoma expressing CD20 by combining with FITC-Her2 and FITC-Rtx, resp. In addn., the activity of anti-FITC CAR T cells could be attenuated by subsequent injections of nonspecific FITC-IgG. Conclusion: These studies highlight an applicability of anti-tag CAR technol. to treat patients with different types of cancers and a possibility to regulate CAR T-cell functions with competing FITC mols.
- 27Ma, J. S.; Kim, J. Y.; Kazane, S. A.; Choi, S. H.; Yun, H. Y.; Kim, M. S.; Rodgers, D. T.; Pugh, H. M.; Singer, O.; Sun, S. B. Versatile strategy for controlling the specificity and activity of engineered T cells. Proc. Natl. Acad. Sci. U.S.A. 2016, 113 (4), E450– E458, DOI: 10.1073/pnas.1524193113Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xns1antA%253D%253D&md5=8a3ca3ccfac0d66d05ec4be93323a970Versatile strategy for controlling the specificity and activity of engineered T cellsMa, Jennifer S. Y.; Kim, Ji Young; Kazane, Stephanie A.; Choi, Sei-hyun; Yun, Hwa Young; Kim, Min Soo; Rodgers, David T.; Pugh, Holly M.; Singer, Oded; Sun, Sophie B.; Fonslow, Bryan R.; Kochenderfer, James N.; Wright, Timothy M.; Schultz, Peter G.; Young, Travis S.; Kim, Chan Hyuk; Cao, YuProceedings of the National Academy of Sciences of the United States of America (2016), 113 (4), E450-E458CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The adoptive transfer of autologous T cells engineered to express a chimeric antigen receptor (CAR) has emerged as a promising cancer therapy. Despite impressive clin. efficacy, the general application of current CAR-T-cell therapy is limited by serious treatment-related toxicities. One approach to improve the safety of CAR-T cells involves making their activation and proliferation dependent upon adaptor mols. that mediate formation of the immunol. synapse between the target cancer cell and T-cell. Here, the authors describe the design and synthesis of structurally defined semisynthetic adaptors the authors refer to as "switch" mols., in which anti-CD19 and anti-CD22 antibody fragments are site-specifically modified with FITC using genetically encoded noncanonical amino acids. This approach allows the precise control over the geometry and stoichiometry of complex formation between CD19- or CD22-expressing cancer cells and a "universal" anti-FITC-directed CAR-T cell. Optimization of this CAR-switch combination results in potent, dose-dependent in vivo antitumor activity in xenograft models. The advantage of being able to titrate CAR-T-cell in vivo activity was further evidenced by reduced in vivo toxicity and the elimination of persistent B-cell aplasia in immune-competent mice. The ability to control CAR-T cell and cancer cell interactions using intermediate switch mols. may expand the scope of engineered T-cell therapy to solid tumors, as well as indications beyond cancer therapy.
- 28Rodgers, D. T.; Mazagova, M.; Hampton, E. N.; Cao, Y.; Ramadoss, N. S.; Hardy, I. R.; Schulman, A.; Du, J.; Wang, F.; Singer, O. Switch-mediated activation and retargeting of CAR-T cells for B-cell malignancies. Proc. Natl. Acad. Sci. U.S.A. 2016, 113 (4), E459– E468, DOI: 10.1073/pnas.1524155113Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xns1antw%253D%253D&md5=a4d4b98acb1877a0394582a6b5bd374cSwitch-mediated activation and retargeting of CAR-T cells for B-cell malignanciesRodgers, David T.; Mazagova, Magdalena; Hampton, Eric N.; Cao, Yu; Ramadoss, Nitya S.; Hardy, Ian R.; Schulman, Andrew; Du, Juanjuan; Wang, Feng; Singer, Oded; Ma, Jennifer; Nunez, Vanessa; Shen, Jiayin; Woods, Ashley K.; Wright, Timothy M.; Schultz, Peter G.; Kim, Chan Hyuk; Young, Travis S.Proceedings of the National Academy of Sciences of the United States of America (2016), 113 (4), E459-E468CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Chimeric antigen receptor T (CAR-T) cell therapy has produced impressive results in clin. trials for B-cell malignancies. However, safety concerns related to the inability to control CAR-T cells once infused into the patient remain a significant challenge. Here the authors report the engineering of recombinant antibody-based bifunctional switches that consist of a tumor antigen-specific Fab mol. engrafted with a peptide neo-epitope, which is bound exclusively by a peptide-specific switchable CAR-T cell (sCAR-T). The switch redirects the activity of the bio-orthogonal sCAR-T cells through the selective formation of immunol. synapses, in which the sCAR-T cell, switch, and target cell interact in a structurally defined and temporally controlled manner. Optimized switches specific for CD19 controlled the activity, tissue-homing, cytokine release, and phenotype of sCAR-T cells in a dose-titratable manner in a Nalm-6 xenograft rodent model of B-cell leukemia. The sCAR-T-cell dosing regimen could be tuned to provide efficacy comparable to the corresponding conventional CART-19, but with lower cytokine levels, thereby offering a method of mitigating cytokine release syndrome in clin. translation. Furthermore, the authors demonstrate that this methodol. is readily adaptable to targeting CD20 on cancer cells using the same sCAR-T cell, suggesting that this approach may be broadly applicable to heterogeneous and resistant tumor populations, as well as other liq. and solid tumor antigens.
- 29Lamers, C. H.; Sleijfer, S.; van Steenbergen, S.; van Elzakker, P.; van Krimpen, B.; Groot, C.; Vulto, A.; den Bakker, M.; Oosterwijk, E.; Debets, R. Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol. Ther. 2013, 21 (4), 904– 912, DOI: 10.1038/mt.2013.17Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXislWhtbw%253D&md5=c6a35182968904a9deb6a7d0890071dcTreatment of Metastatic Renal Cell Carcinoma With CAIX CAR-engineered T cells: Clinical Evaluation and Management of On-target ToxicityLamers, Cor H. J.; Sleijfer, Stefan; van Steenbergen, Sabine; van Elzakker, Pascal; van Krimpen, Brigitte; Groot, Corrien; Vulto, Arnold; den Bakker, Michael; Oosterwijk, Egbert; Debets, Reno; Gratama, Jan W.Molecular Therapy (2013), 21 (4), 904-912CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)Autologous T cells genetically modified to express a chimeric antibody receptor (CAR) against carboxy-anhydrase-IX (CAIX) were administered to 12 patients with CAIX-expressing metastatic renal cell carcinoma (RCC). Patients were treated in three cohorts with a max. of 10 infusions of a total of 0.2 to 2.1 × 109 CAR T cells. CTC grade 2-4 liver enzyme disturbances occurred at the lowest CAR T cell doses, necessitating cessation of treatment in four out of eight patients in cohorts 1 and 2. Examn. of liver biopsies revealed CAIX expression on bile duct epithelium with infiltration of T cells, including CAR T cells. Subsequently four patients were pre-treated with CAIX monoclonal antibody (mAb) G250 to prevent CAR-specific toxicity and showed no liver toxicities and indications for enhanced peripheral T cell persistence. No clin. responses were recorded. This report shows that CAIX-targeting CAR T cells exerted antigen-specific effects in vivo and induced liver toxicity at the lowest dose of 0.2 × 109 T cells applied, illustrating the potency of receptor-modified T cells. We provide in-patient proof that the obsd. "on-target" toxicity is antigen-directed and can be prevented by blocking antigenic sites in off-tumor organs and allowing higher T cell doses.
- 30Morgan, R. A.; Yang, J. C.; Kitano, M.; Dudley, M. E.; Laurencot, C. M.; Rosenberg, S. A. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 2010, 18 (4), 843– 851, DOI: 10.1038/mt.2010.24Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXitlaitb4%253D&md5=fdf1fd771d8ce6bb9c83f46e58d89db6Case Report of a Serious Adverse Event Following the Administration of T Cells Transduced With a Chimeric Antigen Receptor Recognizing ERBB2Morgan, Richard A.; Yang, James C.; Kitano, Mio; Dudley, Mark E.; Laurencot, Carolyn M.; Rosenberg, Steven A.Molecular Therapy (2010), 18 (4), 843-851CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)In an attempt to treat cancer patients with ERBB2 overexpressing tumors, we developed a chimeric antigen receptor (CAR) based on the widely used humanized monoclonal antibody (mAb) Trastuzumab (Herceptin). An optimized CAR vector contg. CD28, 4-1BB, and CD3ζ signaling moieties was assembled in a γ-retroviral vector and used to transduce autologous peripheral blood lymphocytes (PBLs) from a patient with colon cancer metastatic to the lungs and liver, refractory to multiple std. treatments. The gene transfer efficiency into autologous T cells was 79% CAR+ in CD3+ cells and these cells demonstrated high-specific reactivity in in vitro coculture assays. Following completion of nonmyeloablative conditioning, the patient received 1010 cells i.v. Within 15 min after cell infusion the patient experienced respiratory distress, and displayed a dramatic pulmonary infiltrate on chest X-ray. She was intubated and despite intensive medical intervention the patient died 5 days after treatment. Serum samples after cell infusion showed marked increases in interferon-γ (IFN-γ), granulocyte macrophage-colony stimulating factor (GM-CSF), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-10, consistent with a cytokine storm. We speculate that the large no. of administered cells localized to the lung immediately following infusion and were triggered to release cytokine by the recognition of low levels of ERBB2 on lung epithelial cells.
- 31Gardner, L.; Deiters, A. Light-controlled synthetic gene circuits. Curr. Opin. Chem. Biol. 2012, 16 (3–4), 292– 299, DOI: 10.1016/j.cbpa.2012.04.010Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XnslSitro%253D&md5=1e2bc100aebe766aed22d2937bba8f47Light-controlled synthetic gene circuitsGardner, Laura; Deiters, AlexanderCurrent Opinion in Chemical Biology (2012), 16 (3-4), 292-299CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. Highly complex synthetic gene circuits have been engineered in living organisms to develop systems with new biol. properties. A precise trigger to activate or deactivate these complex systems is desired in order to tightly control different parts of a synthetic or natural network. Light represents an excellent tool to achieve this goal as it can be regulated in timing, location, intensity, and wavelength, which allows for precise spatiotemporal control over genetic circuits. Recently, light has been used as a trigger to control the biol. function of small mols., oligonucleotides, and proteins involved as parts in gene circuits. Light activation has enabled the construction of unique systems in living organisms such as band-pass filters and edge-detectors in bacterial cells. Addnl., light also allows for the regulation of intermediate steps of complex dynamic pathways in mammalian cells such as those involved in kinase networks. Herein we describe recent advancements in the area of light-controlled synthetic networks.
From NLM Medline
- 32Ankenbruck, N.; Courtney, T.; Naro, Y.; Deiters, A. Optochemical Control of Biological Processes in Cells and Animals. Angew. Chem., Int. Ed. Engl. 2018, 57 (11), 2768– 2798, DOI: 10.1002/anie.201700171Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1crovFGnsg%253D%253D&md5=8f1ab7383a0629b0ed87b58d47ad0727Optochemical Control of Biological Processes in Cells and AnimalsAnkenbruck Nicholas; Courtney Taylor; Naro Yuta; Deiters AlexanderAngewandte Chemie (International ed. in English) (2018), 57 (11), 2768-2798 ISSN:.Biological processes are naturally regulated with high spatial and temporal control, as is perhaps most evident in metazoan embryogenesis. Chemical tools have been extensively utilized in cell and developmental biology to investigate cellular processes, and conditional control methods have expanded applications of these technologies toward resolving complex biological questions. Light represents an excellent external trigger since it can be controlled with very high spatial and temporal precision. To this end, several optically regulated tools have been developed and applied to living systems. In this review we discuss recent developments of optochemical tools, including small molecules, peptides, proteins, and nucleic acids that can be irreversibly or reversibly controlled through light irradiation, with a focus on applications in cells and animals.
From NLM Medline
- 33Davies, S.; Stenton, B. J.; Bernardes, G. J. L. Bioorthogonal Decaging Reactions for Targeted Drug Activation. Chimia 2018, 72 (11), 771– 776, DOI: 10.2533/chimia.2018.771Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvVaqsLs%253D&md5=21cc53fef93345d64f1c9351b9c83debBioorthogonal decaging reactions for targeted drug activationDavies, Sarah; Stenton, Benjamin J.; Bernardes, Goncalo J. L.Chimia (2018), 72 (11), 771-776CODEN: CHIMAD ISSN:. (Swiss Chemical Society)Bioorthogonal decaging reactions are highly selective transformations which involve the cleavage of a protecting group from a mol. of interest. Decaging reactions can be classified into subgroups depending on the nature of the trigger; they can be photo-, metal- or small mol.-triggered. Due to their highly selective and biocompatible nature, they can be carried out in living systems as they do not interfere with any endogenous processes. This gain-of-function allows controlled activation of proteins and release of fluorophores and drugs in vivo. Although there are many examples of fluorophore/protein release, this review focuses on the application of bioorthogonal decaging reactions for targeted drug activation. One strategy for targeted drug delivery is tissue-selective activation of prodrugs and antibody-drug conjugates (ADCs). Bioorthogonal decaging provides a highly selective, controllable method for activating prodrugs and ADCs, reducing toxicity due to the off-target drug release that occurs in endogenous activation strategies. Here we focus on the development of bifunctional linkers that enable studies of bioorthogonal chem. for activation of ADCs.
From NLM Medline
- 34van der Gracht, A. M. F.; de Geus, M. A. R.; Camps, M. G. M.; Ruckwardt, T. J.; Sarris, A. J. C.; Bremmers, J.; Maurits, E.; Pawlak, J. B.; Posthoorn, M. M.; Bonger, K. M. Chemical Control over T-Cell Activation in Vivo Using Deprotection of trans-Cyclooctene-Modified Epitopes. ACS Chem. Biol. 2018, 13 (6), 1569– 1576, DOI: 10.1021/acschembio.8b00155Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MfgsVKrtA%253D%253D&md5=e2ae7a8dc3a10d411afcd5873a596eadChemical Control over T-Cell Activation in Vivo Using Deprotection of trans-Cyclooctene-Modified Epitopesvan der Gracht Anouk M F; de Geus Mark A R; Sarris Alexi J C; Bremmers Jessica; Maurits Elmer; Pawlak Joanna B; Posthoorn Michelle M; Filippov Dmitri V; Overkleeft Herman S; van Kasteren Sander I; Camps Marcel G M; Ossendorp Ferry; Ruckwardt Tracy J; Bonger Kimberly M; Robillard Marc SACS chemical biology (2018), 13 (6), 1569-1576 ISSN:.Activation of a cytotoxic T-cell is a complex multistep process, and tools to study the molecular events and their dynamics that result in T-cell activation in situ and in vivo are scarce. Here, we report the design and use of conditional epitopes for time-controlled T-cell activation in vivo. We show that trans-cyclooctene-protected SIINFEKL (with the lysine amine masked) is unable to elicit the T-cell response characteristic for the free SIINFEKL epitope. Epitope uncaging by means of an inverse-electron demand Diels-Alder (IEDDA) event restored T-cell activation and provided temporal control of T-cell proliferation in vivo.
From NLM Medline
- 35Li, J.; Chen, P. R. Development and application of bond cleavage reactions in bioorthogonal chemistry. Nat. Chem. Biol. 2016, 12 (3), 129– 137, DOI: 10.1038/nchembio.2024Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XivVCis74%253D&md5=6b563f0c618e43f97bf4cea26c060451Development and application of bond cleavage reactions in bioorthogonal chemistryLi, Jie; Chen, Peng R.Nature Chemical Biology (2016), 12 (3), 129-137CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)A review. Bioorthogonal chem. reactions are a thriving area of chem. research in recent years as an unprecedented technique to dissect native biol. processes through chem.-enabled strategies. However, current concepts of bioorthogonal chem. have largely centered on bond formation reactions between two mutually reactive bioorthogonal handles. Recently, in a reverse strategy, a collection of bond cleavage reactions has emerged with excellent biocompatibility. These reactions have expanded the bioorthogonal chem. repertoire, enabling an array of exciting new biol. applications that range from the chem. controlled spatial and temporal activation of intracellular proteins and small-mol. drugs to the direct manipulation of intact cells under physiol. conditions. Here the authors highlight the development and applications of these bioorthogonal cleavage reactions. Furthermore, the authors lay out challenges and propose future directions along this appealing avenue of research.
From NLM Medline
- 36Scinto, S. L.; Bilodeau, D. A.; Hincapie, R.; Lee, W.; Nguyen, S. S.; Xu, M.; Am Ende, C. W.; Finn, M. G.; Lang, K.; Lin, Q. Bioorthogonal chemistry. Nat. Rev. Methods Primers 2021, 1, 30, DOI: 10.1038/s43586-021-00028-zGoogle Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XjsVGgu7k%253D&md5=64eb15e12374df94c7b27bc4c9ef2accBioorthogonal chemistryScinto, Samuel L.; Bilodeau, Didier A.; Hincapie, Robert; Lee, Wankyu; Nguyen, Sean S.; Xu, Minghao; am Ende, Christopher W.; Finn, M. G.; Lang, Kathrin; Lin, Qing; Pezacki, John Paul; Prescher, Jennifer A.; Robillard, Marc S.; Fox, Joseph M.Nature Reviews Methods Primers (2021), 1 (1), 30CODEN: NRMPAT; ISSN:2662-8449. (Nature Portfolio)A review. Bioorthogonal chem. represents a class of high-yielding chem. reactions that proceed rapidly and selectively in biol. environments without side reactions towards endogenous functional groups. Rooted in the principles of phys. org. chem., bioorthogonal reactions are intrinsically selective transformations not commonly found in biol. Key reactions include native chem. ligation and the Staudinger ligation, copper-catalyzed azide-alkyne cycloaddn., strain-promoted [3 + 2] reactions, tetrazine ligation, metal-catalyzed coupling reactions, oxime and hydrazone ligations as well as photoinducible bioorthogonal reactions. Bioorthogonal chem. has significant overlap with the broader field of click chem. - high-yielding reactions that are wide in scope and simple to perform, as recently exemplified by sulfuryl fluoride exchange chem. The underlying mechanisms of these transformations and their optimal conditions are described in this Primer, followed by discussion of how bioorthogonal chem. has become essential to the fields of biomedical imaging, medicinal chem., protein synthesis, polymer science, materials science and surface science. The applications of bioorthogonal chem. are diverse and include genetic code expansion and metabolic engineering, drug target identification, antibody-drug conjugation and drug delivery. This Primer describes stds. for reproducibility and data deposition, outlines how current limitations are driving new research directions and discusses new opportunities for applying bioorthogonal chem. to emerging problems in biol. and biomedicine.
- 37Tu, J.; Xu, M.; Franzini, R. M. Dissociative Bioorthogonal Reactions. ChemBioChem 2019, 20 (13), 1615– 1627, DOI: 10.1002/cbic.201800810Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXotFamsbY%253D&md5=e9277eac67c6092b293c936477e47467Dissociative Bioorthogonal ReactionsTu, Julian; Xu, Minghao; Franzini, Raphael M.ChemBioChem (2019), 20 (13), 1615-1627CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)Bioorthogonal reactions that proceed readily under physiol. conditions without interference from biomols. have found widespread application in the life sciences. Complementary to the bioorthogonal reactions that ligate two mols., reactions that release a mol. or cleave a linker are increasingly attracting interest. Such dissociative bioorthogonal reactions have a broad spectrum of uses, for example, in controlling bio-macromol. activity, in drug delivery, and in diagnostic assays. This review article summarizes the developed bioorthogonal reactions linked to a release step, outlines representative areas of the applications of such reactions, and discusses aspects that require further improvement.
From NLM Medline
- 38Messmann, H.; Endlicher, E.; Freunek, G.; Rummele, P.; Scholmerich, J.; Knuchel, R. Fluorescence endoscopy for the detection of low and high grade dysplasia in ulcerative colitis using systemic or local 5-aminolaevulinic acid sensitisation. Gut 2003, 52 (7), 1003– 1007, DOI: 10.1136/gut.52.7.1003Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3s3nsFantQ%253D%253D&md5=19bd79f280aeb54b5a1a4dac1f95490dFluorescence endoscopy for the detection of low and high grade dysplasia in ulcerative colitis using systemic or local 5-aminolaevulinic acid sensitisationMessmann H; Endlicher E; Freunek G; Rummele P; Scholmerich J; Knuchel RGut (2003), 52 (7), 1003-7 ISSN:0017-5749.BACKGROUND AND AIMS: Longstanding ulcerative colitis (UC), especially in the presence of epithelial dysplasia, is associated with an increased risk of developing cancer. As dysplasia is not visible during routine endoscopy, at least 40-50 random biopsies in four quadrants every 10 cm are recommended. Fluorescence endoscopy after sensitisation with 5-aminolaevulinic acid (5-ALA) was assessed for the detection of dysplasia in ulcerative colitis by taking optical guided biopsies. 5-ALA is converted intracellularly into the sensitiser protoporphyrin IX which accumulates selectively in neoplastic tissue allowing the detection of dysplasia by typical red fluorescence after illumination with blue light. METHODS: In 37 patients with UC, 54 examinations were performed with fluorescence endoscopy after oral (20 mg/kg) or local (either with an enema or by spraying the mucosa with a catheter) sensitisation with 5-ALA. A total of 481 biopsies of red fluorescent (n=218) and non-fluorescent (n=263) areas of the colonic mucosa were taken. RESULTS: Forty two biopsies in 12 patients revealed either low grade (n=40) or high grade (n=2) dysplasia. Sensitivity of fluorescence for dysplastic lesions was excellent and ranged from 87% (95% confidence interval (CI) 0.73-1.00) to 100% (95% CI 1.00-1.00) after local sensitisation, in contrast with only 43% (95% CI 0.17-0.69) after systemic administration. Specificity did not differ for both forms of local sensitisation (enema 51% (95% CI 0.44-0.57) and spray catheter 62% (95% CI 0.51-0.73)); after systemic sensitisation specificity was 73% (95% CI 0.69-0.83). Negative predictive values of non-fluorescent mucosa for exclusion of dysplasia were very high; 89% after systemic sensitisation and 98-100% after local sensitisation. Positive predictive values were 13% and 14% after local sensitisation with enema and spray catheter, and 21% after oral sensitisation with 20 mg/kg ALA. The overall number of biopsies per examination was less than five from fluorescent positive areas. CONCLUSION: Fluorescence endoscopy after 5-ALA sensitisation is a possible tool to visualise dysplastic lesions in ulcerative colitis using 5-ALA sensitisation. Local sensitisation is a promising alternative approach compared with systemic administration of 5-ALA. A randomised controlled study is now indicated to compare the efficacy of endoscopic fluorescence detection with the standard technique of four quadrant random biopsies.
- 39Georgianna, W. E.; Lusic, H.; McIver, A. L.; Deiters, A. Photocleavable polyethylene glycol for the light-regulation of protein function. Bioconjugate Chem. 2010, 21 (8), 1404– 1407, DOI: 10.1021/bc100084nGoogle Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptFGgur4%253D&md5=117f49d83b85ea19b2f735335ac18184Photocleavable Polyethylene Glycol for the Light-Regulation of Protein FunctionGeorgianna, Wesleigh E.; Lusic, Hrvoje; McIver, Andrew L.; Deiters, AlexanderBioconjugate Chemistry (2010), 21 (8), 1404-1407CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)PEGylation is commonly employed to enhance the pharmacokinetic properties of proteins, but it can interfere with natural protein function. Protein activity can thus be abrogated through PEGylation, and a controllable means to remove the polyethylene glycol (PEG) group from the protein is desirable. As such, light affords a unique control over biomols. through the application of photosensitive groups. Herein, the authors report the synthesis of a photocleavable PEG reagent (PhotoPEG) and its application to the light-regulation of enzyme activity.
From NLM Medline
- 40Govan, J. M.; McIver, A. L.; Deiters, A. Stabilization and photochemical regulation of antisense agents through PEGylation. Bioconjugate Chem. 2011, 22 (10), 2136– 2142, DOI: 10.1021/bc200411nGoogle Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1ChtLfO&md5=b0a42ab4adfaee28ac04e19c4ebf2679Stabilization and Photochemical Regulation of Antisense Agents through PEGylationGovan, Jeane M.; McIver, Andrew L.; Deiters, AlexanderBioconjugate Chemistry (2011), 22 (10), 2136-2142CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Oligonucleotides are effective tools for the regulation of gene expression in cell culture and model organisms, most importantly through antisense mechanisms. Due to the inherent instability of DNA antisense agents, various modifications have been introduced to increase the efficacy of oligonucleotides, including phosphorothioate DNA, locked nucleic acids, peptide nucleic acids, and others. Here, we present antisense agent stabilization through conjugation of a poly(ethylene glycol) (PEG) group to a DNA oligonucleotide. By employing a photocleavable linker between the PEG group and the antisense agent, we were able to achieve light-induced deactivation of antisense activity. The bioconjugated PEG group provides stability to the DNA antisense agent without affecting its native function of silencing gene expression via RNase H-catalyzed mRNA degrdn. Once irradiated with UV light of 365 nm, the PEG group is cleaved from the antisense agent leaving the DNA unprotected and open for degrdn. by endogenous nucleases, thereby restoring gene expression. By using a photocleavable PEG group (PhotoPEG), antisense activity can be regulated with high spatial and temporal resoln., paving the way for precise regulation of gene expression in biol. systems.
From NLM Medline
- 41Govan, J. M.; Uprety, R.; Thomas, M.; Lusic, H.; Lively, M. O.; Deiters, A. Cellular delivery and photochemical activation of antisense agents through a nucleobase caging strategy. ACS Chem. Biol. 2013, 8 (10), 2272– 2282, DOI: 10.1021/cb400293eGoogle Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1Srtr7P&md5=d7442889bc061661f09617e5656092cbCellular Delivery and Photochemical Activation of Antisense Agents through a Nucleobase Caging StrategyGovan, Jeane M.; Uprety, Rajendra; Thomas, Meryl; Lusic, Hrvoje; Lively, Mark O.; Deiters, AlexanderACS Chemical Biology (2013), 8 (10), 2272-2282CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Antisense oligonucleotides are powerful tools to regulate gene expression in cells and model organisms. However, a transfection or microinjection is typically needed for efficient delivery of the antisense agent. We report the conjugation of multiple HIV TAT peptides to a hairpin-protected antisense agent through a light-cleavable nucleobase caging group. This conjugation allows for the facile delivery of the antisense agent without a transfection reagent, and photochem. activation offers precise control over gene expression. The developed approach is highly modular, as demonstrated by the conjugation of folic acid to the caged antisense agent. This enabled targeted cell delivery through cell-surface folate receptors followed by photochem. triggering of antisense activity. Importantly, the presented strategy delivers native oligonucleotides after light-activation, devoid of any delivery functionalities or modifications that could otherwise impair their antisense activity.
From NLM Medline
- 42Yamazoe, S.; Liu, Q.; McQuade, L. E.; Deiters, A.; Chen, J. K. Sequential gene silencing using wavelength-selective caged morpholino oligonucleotides. Angew. Chem., Int. Ed. Engl. 2014, 53 (38), 10114– 10118, DOI: 10.1002/anie.201405355Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2M%252FisVyitw%253D%253D&md5=3b0e34a24cb4f5fd53ddff365df002b5Sequential gene silencing using wavelength-selective caged morpholino oligonucleotidesYamazoe Sayumi; Liu Qingyang; McQuade Lindsey E; Deiters Alexander; Chen James KAngewandte Chemie (International ed. in English) (2014), 53 (38), 10114-8 ISSN:.Spectrally differentiated caged morpholino oligonucleotides (cMOs) and wavelength-selective illumination have been used to sequentially inactivate organismal gene function. The efficacy of these reverse-genetic chemical probes has been demonstrated in zebrafish embryos, and these reagents have been employed to examine the mechanisms of mesoderm patterning.
From NLM Medline
- 43Brown, W.; Wesalo, J.; Tsang, M.; Deiters, A. Engineering Small Molecule Switches of Protein Function in Zebrafish Embryos. J. Am. Chem. Soc. 2023, 145 (4), 2395– 2403, DOI: 10.1021/jacs.2c11366Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXhsVeit70%253D&md5=5eca14fb80e49c840a9f63e3da5217a9Engineering Small Molecule Switches of Protein Function in Zebrafish EmbryosBrown, Wes; Wesalo, Joshua; Tsang, Michael; Deiters, AlexanderJournal of the American Chemical Society (2023), 145 (4), 2395-2403CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Precise temporally regulated protein function directs the highly complex processes that make up embryo development. The zebrafish embryo is an excellent model organism to study development, and conditional control over enzymic activity is desirable to target chem. intervention to specific developmental events and to investigate biol. mechanisms. Surprisingly, however, few generally applicable small mol. switches of protein function exist in zebrafish. Genetic code expansion allows for site-specific incorporation of an unnatural amino acids into proteins that contain caging groups that are removed through addn. of small mol. triggers such as phosphines or tetrazines. This broadly applicable control of protein function was applied to activate several enzymes, including a GTPase and a protease, with temporal precision in zebrafish embryos. Simple addn. of the small mol. trigger to the media produces robust and tunable protein activation, which was used to gain insight into the development of a congenital heart defect from a RASopathy mutant of NRAS and to control DNA and protein cleavage events catalyzed by a viral recombinase and a viral protease, resp.
From NLM Medline
- 44Darrah, K.; Wesalo, J.; Lukasak, B.; Tsang, M.; Chen, J. K.; Deiters, A. Small Molecule Control of Morpholino Antisense Oligonucleotide Function through Staudinger Reduction. J. Am. Chem. Soc. 2021, 143 (44), 18665– 18671, DOI: 10.1021/jacs.1c08723Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitlajur%252FF&md5=2d697c181ecf19a44cc5681fb079b47bSmall Molecule Control of Morpholino Antisense Oligonucleotide Function through Staudinger ReductionDarrah, Kristie; Wesalo, Joshua; Lukasak, Bradley; Tsang, Michael; Chen, James K.; Deiters, AlexanderJournal of the American Chemical Society (2021), 143 (44), 18665-18671CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Conditionally activated, caged morpholino antisense agents (cMOs) are tools that enable the temporal and spatial investigation of gene expression, regulation, and function during embryonic development. Cyclic MOs are conformationally gated oligonucleotide analogs that do not block gene expression until they are linearized through the application of an external trigger, such as light or enzyme activity. Here, we describe the first examples of small mol.-responsive cMOs, which undergo rapid and efficient decaging via a Staudinger redn. This is enabled by a highly flexible linker design that offers opportunities for the installation of chem. activated, self-immolative motifs. We synthesized cyclic cMOs against two distinct, developmentally relevant genes and demonstrated phosphine-triggered knockdown of gene expression in zebrafish embryos. This represents the first report of a small mol.-triggered antisense agent for gene knockdown, adding another bioorthogonal entry to the growing arsenal of gene knockdown tools.
From NLM Medline
- 45Lukasak, B.; Morihiro, K.; Deiters, A. Aryl Azides as Phosphine-Activated Switches for Small Molecule Function. Sci. Rep. 2019, 9 (1), 1470, DOI: 10.1038/s41598-018-37023-6Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cjpvV2mtg%253D%253D&md5=4b9da691d5a6aa1655202d35493630f4Aryl Azides as Phosphine-Activated Switches for Small Molecule FunctionLukasak Bradley; Morihiro Kunihiko; Deiters AlexanderScientific reports (2019), 9 (1), 1470 ISSN:.Engineered small molecule triggers are important tools for the control and investigation of biological processes, in particular protein function. Staudinger reductions of aryl azides to amines through the use of phosphines can trigger an elimination reaction, and thereby activation of a functional molecule, if an appropriately positioned leaving group is present. We conducted detailed investigations of the effect of aryl azide and phosphine structure on both the mechanism and kinetics of these reaction-induced eliminations and identified phosphine/azide pairs that enable complete activation within minutes under physiologically relevant conditions.
From NLM Medline
- 46Wesalo, J. S.; Luo, J.; Morihiro, K.; Liu, J.; Deiters, A. Phosphine-Activated Lysine Analogues for Fast Chemical Control of Protein Subcellular Localization and Protein SUMOylation. ChemBioChem 2020, 21 (1–2), 141– 148, DOI: 10.1002/cbic.201900464Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitVCkt73L&md5=c43a9a16ad9d11ecacbeb5225570547aPhosphine-Activated Lysine Analogues for Fast Chemical Control of Protein Subcellular Localization and Protein SUMOylationWesalo, Joshua S.; Luo, Ji; Morihiro, Kunihiko; Liu, Jihe; Deiters, AlexanderChemBioChem (2020), 21 (1-2), 141-148CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)The Staudinger redn. and its variants have exceptional compatibility with live cells but can be limited by slow kinetics. Herein we report new small-mol. triggers that turn on proteins through a Staudinger redn./self-immolation cascade with substantially improved kinetics and yields. We achieved this through site-specific incorporation of a new set of azidobenzyloxycarbonyl lysine derivs. in mammalian cells. This approach allowed us to activate proteins by adding a nontoxic, bioorthogonal phosphine trigger. We applied this methodol. to control a post-translational modification (SUMOylation) in live cells, using native modification machinery. This work significantly improves the rate, yield, and tunability of the Staudinger redn.-based activation, paving the way for its application in other proteins and organisms.
From NLM Medline
- 47Lim, K. H.; Huang, H.; Pralle, A.; Park, S. Stable, high-affinity streptavidin monomer for protein labeling and monovalent biotin detection. Biotechnol. Bioeng. 2013, 110 (1), 57– 67, DOI: 10.1002/bit.24605Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFKlsbjK&md5=62b50d6b861c5cf02ffca311e39acb43Stable, high-affinity streptavidin monomer for protein labeling and monovalent biotin detectionLim, Kok Hong; Huang, Heng; Pralle, Arnd; Park, SheldonBiotechnology and Bioengineering (2013), 110 (1), 57-67CODEN: BIBIAU; ISSN:0006-3592. (John Wiley & Sons, Inc.)The coupling between the quaternary structure, stability and function of streptavidin makes it difficult to engineer a stable, high affinity monomer for biotechnol. applications. For example, the binding pocket of streptavidin tetramer is comprised of residues from multiple subunits, which cannot be replicated in a single domain protein. However, rhizavidin from Rhizobium etli was recently shown to bind biotin with high affinity as a dimer without the hydrophobic tryptophan lid donated by an adjacent subunit. In particular, the binding site of rhizavidin uses residues from a single subunit to interact with bound biotin. The authors therefore postulated that replacing the binding site residues of streptavidin monomer with corresponding rhizavidin residues would lead to the design of a high affinity monomer useful for biotechnol. applications. Here, the authors report the construction and characterization of a structural monomer, mSA, which combines the streptavidin and rhizavidin sequences to achieve optimized biophys. properties. First, the biotin affinity of mSA (Kd = 2.8 nM) is the highest among nontetrameric streptavidin, allowing sensitive monovalent detection of biotinylated ligands. The monomer also has significantly higher stability (Tm = 59.8°) and soly. than all other previously engineered monomers to ensure the mol. remains folded and functional during its application. Using fluorescence correlation spectroscopy, mSA binds biotinylated targets as a monomer. Also the mol. can be used as a genetic tag to introduce biotin binding capability to a heterologous protein. For example, recombinantly fusing the monomer to a cell surface receptor allows direct labeling and imaging of transfected cells using biotinylated fluorophores. A stable and functional streptavidin monomer, such as mSA, should be a useful reagent for designing novel detection systems based on monovalent biotin interaction.
- 48Begovic, M.; Herberman, R. B.; Gorelik, E. Ultraviolet light-induced increase in tumor cell susceptibility to TNF-dependent and TNF-independent natural cell-mediated cytotoxicity. Cell. Immunol. 1991, 138 (2), 349– 359, DOI: 10.1016/0008-8749(91)90159-9Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38XjslWjsA%253D%253D&md5=d2030dbd85b108b7f1081be393a6c4baUltraviolet light-induced increase in tumor cell susceptibility to TNF-dependent and TNF-independent natural cell-mediated cytotoxicityBegovic, Mirsada; Herberman, Ronald B.; Gorelik, ElieserCellular Immunology (1991), 138 (2), 349-59CODEN: CLIMB8; ISSN:0008-8749.Anal. of the effector cells involved in lysis of MCA102 tumor cells was performed by comparing the cytotoxicity of normal spleen cells which mediated both NK and natural cell (NC) cell activity with (a) normal spleen cells in which NC activity was neutralized by anti-TNF Abs (NK+,NC-), (b) NK-depleted or NK-deficient spleen cells (NK-,NC+), and (c) NK-deficient or -depleted spleen cells with NC activity neutralized by anti-TNF Abs (NK-,NC-). Lysis of the original MCA102 tumor cells was relatively low and was mediated by NC cells. UV irradn. increased MCA102 tumor cell sensitivity to lysis by both NK and NC cells. Anal. of the mechanisms involved in UV-induced NK sensitivity revealed that UV irradn. increased tumor cell susceptibility to lytic NK-derived granules. NC sensitivity of MCA102UV tumor cells was assocd. with their increase in sensitivity to TNF and selection of MCA102UV cells for resistance to rTNF resulted in a decrease in their susceptibility to NC cells. To det. how fast UV-induced sensitivity to NCMC and rTNF can be established, 51Cr-labeled MCA102 cells were irradiated in vitro with 38-304 J/m2 of UVC light and their sensitivity to lysis by spleen cells and rTNF was tested immediately in an 18-h cytotoxicity assay. UV treatment with the same doses was repeated 12 days later. The data obtained showed that tumor cell sensitivity to NCMC and TNF appeared shortly after UV irradn., was stable, and was further substantially augmented by the 2nd round of UV treatment. Thus, in vitro UV irradn. of tumor cells could be an effective modulator of tumor cell sensitivity to TNF-dependent and TNF-independent cell-mediated cytotoxicity.
From NLM Medline
- 49Begovic, M.; Herberman, R. B.; Gorelik, E. Effect of UV light on tumor cell sensitivity to NK and NC cell-mediated lysis. Nat. Immun 1993, 12 (4–5), 250– 266Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXhvVKhtbs%253D&md5=b781014a1179ace46515fd3b502198c3Effect of UV light on tumor cell sensitivity to NK and NC cell-mediated lysisBegovic, Mirsada; Herberman, Ronald B.; Gorelik, ElieserNatural Immunity (1993), 12 (4-5), 250-66CODEN: NAIMEL; ISSN:1018-8916.The effect of UV light irradn. on the immunobiol. properties of murine tumor cells was studied. In vitro irradn. of MCA 102 and MCA 105 fibrosarcomas with a short-wavelength UVC light rendered them highly immunogenic and sensitive to natural cell-mediated cytotoxicity (NCMC). Anal. of the effector cells involved in NCMC revealed that UV irradn. stably increased tumor cell sensitivity to both NK and NC cell lysis. Studies of the mechanisms responsible for increased sensitivity to NK cells indicate that UV treatment did not affect tumor-cell recognition by NK cells but increased their susceptibility to NK-derived lytic granules. Augmentation of UV-treated tumor cell sensitivity to NC cell-mediated lysis was found to be due to their increase in sensitivity to the effector-cell-released TNF. In parallel, UV-treated cells showed high sensitivity to human recombinant TNF whereas untreated parental cells were resistant to rTNF. UV irradn. did not affect rTNF binding, internalization and degrdn. but increased tumor cell vulnerability to TNF-induced DNA fragmentation. Thus, UV light appears as a potent modulator of tumor cell sensitivity to T cell-and natural cell-mediated immunity.
From NLM Medline.
- 50Gorelik, E.; Begovic, M.; Duty, L.; Herberman, R. B. Effect of ultraviolet irradiation on MCA102 tumor cell immunogenicity and sensitivity to tumor necrosis factor. Cancer Res. 1991, 51 (5), 1521– 1528Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXhtl2ju7c%253D&md5=b3d96f18264900670e3e06d0f8710c03Effect of ultraviolet irradiation on MCA102 tumor cell immunogenicity and sensitivity to tumor necrosis factorGorelik, Elieser; Begovic, Mirsada; Duty, Lisa; Herberman, Ronald B.Cancer Research (1991), 51 (5), 1521-8CODEN: CNREA8; ISSN:0008-5472.The ability of UV irradn. to induce immunogenicity of the nonimmunogenic major histocompatibility complex-neg. MCA102 fibrosarcoma was studied. In parallel, the effect of short wavelength UVC light on the sensitivity of tumor cells to natural cell-mediated cytotoxicity and tumor necrosis factor (TNF) was also investigated. MCA102 fibrosarcoma cells were irradiated in vitro twice with UVC light (610 and 457 J/m2). UV treatment changed tumor cell morphol. and increased their in vitro rate of proliferation. However, after inoculation of 1 × 105 to 2 × 106 MCA102UV cells into C57BL/6 mice, growth of these cells was completely prevented. Lyt2.2 and not L3T4 lymphocytes were responsible for the rejection of these tumor cells. After a single dose of UV treatment, tumor growth in C57BL/6 mice was inhibited, particularly with lines irradiated at the highest doses (610 or 457 J/m2). After a 2nd round of irradn., tumor cells became more immunogenic, and the level of tumor growth inhibition increased with higher doses of UV irradn. Thus, cells irradiated twice with 610 and 457 J/m2 became rejectable in all immunocompetent C57BL/6 mice. The increase in tumor cell immunogenicity induced by UV light was not assocd. with the appearance of Class I H-2 antigens. In parallel with the induction of tumor cell immunogenicity, UV irradn. made tumor cells more sensitive to natural cell-mediated cytotoxicity and to lysis by TNF. An increase in sensitivity to natural cell-mediated cytotoxicity and TNF was obsd. after single or double doses (152-610 J/m2) of UV irradn. Thus, UV irradn. appears to be an effective modality for altering tumor cell immunobiol. properties, including increased tumor cell sensitivity to T-cell and/or natural cell-mediated immunity.
From NLM Medline.
- 51Jutz, S.; Leitner, J.; Schmetterer, K.; Doel-Perez, I.; Majdic, O.; Grabmeier-Pfistershammer, K.; Paster, W.; Huppa, J. B.; Steinberger, P. Assessment of costimulation and coinhibition in a triple parameter T cell reporter line: Simultaneous measurement of NF-κB, NFAT and AP-1. J. Immunol. Methods 2016, 430, 10– 20, DOI: 10.1016/j.jim.2016.01.007Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVKhsb4%253D&md5=3c84198664801ba14aecfb27ba91164cAssessment of costimulation and coinhibition in a triple parameter T cell reporter line: Simultaneous measurement of NF-κB, NFAT and AP-1Jutz, Sabrina; Leitner, Judith; Schmetterer, Klaus; Doel-Perez, Iago; Majdic, Otto; Grabmeier-Pfistershammer, Katharina; Paster, Wolfgang; Huppa, Johannes B.; Steinberger, PeterJournal of Immunological Methods (2016), 430 (), 10-20CODEN: JIMMBG; ISSN:0022-1759. (Elsevier B.V.)Engagement of the T cell receptor complex reprograms T cells for proliferation, cytokine prodn. and differentiation towards effector cells. This process depends on activating costimulatory signals and is counteracted by coinhibitory mols. Three transcription factors, namely NF-κB, NFAT and AP-1, have a major role in inducing the transcriptional program that is required for T cell activation and differentiation. Here we describe the generation of a triple parameter reporter based on the human Jurkat T cell line, where response elements for NF-κB, NFAT and AP-1 drive the expression of the fluorescent proteins CFP, eGFP and mCherry, resp. The emission spectra of these proteins allow simultaneous assessment of NF-κB, NFAT and AP-1 activity in response to stimulation. Ligation of the TCR complex induced moderate reporter activity, which was strongly enhanced upon coengagement of the costimulatory receptors CD2 or CD28. Moreover, we have generated and tested triple parameter reporter cells that harbor costimulatory and inhibitory receptors not endogenously expressed in the Jurkat cells. In these expts. we could show that engagement of the costimulatory mol. 4-1BB enhances NF-κB and AP-1 activity, whereas coinhibition via PD-1 or BTLA strongly reduced the activation of NF-κB and NFAT. Engagement of BTLA significantly inhibited AP-1, whereas PD-1 had little effect on the activation of this transcription factor. Our triple parameter reporter T cell line is an excellent tool to assess the effect of costimulatory and coinhibitory receptors on NF-κB, NFAT and AP-1 activity and has a wide range of applications beyond the evaluation of costimulatory pathways.
From NLM Medline
- 52Zhang, L.; Morgan, R. A.; Beane, J. D.; Zheng, Z.; Dudley, M. E.; Kassim, S. H.; Nahvi, A. V.; Ngo, L. T.; Sherry, R. M.; Phan, G. Q. Tumor-infiltrating lymphocytes genetically engineered with an inducible gene encoding interleukin-12 for the immunotherapy of metastatic melanoma. Clin. Cancer Res. 2015, 21 (10), 2278– 2288, DOI: 10.1158/1078-0432.CCR-14-2085Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXoslOktbk%253D&md5=7fb2e4d9251f1e3af67d1e0f826a6eefTumor-Infiltrating Lymphocytes Genetically Engineered with an Inducible Gene Encoding Interleukin-12 for the Immunotherapy of Metastatic MelanomaZhang, Ling; Morgan, Richard A.; Beane, Joal D.; Zheng, Zhili; Dudley, Mark E.; Kassim, Sadik H.; Nahvi, Azam V.; Ngo, Lien T.; Sherry, Richard M.; Phan, Giao Q.; Hughes, Marybeth S.; Kammula, Udai S.; Feldman, Steven A.; Toomey, Mary Ann; Kerkar, Sid P.; Restifo, Nicholas P.; Yang, James C.; Rosenberg, Steven A.Clinical Cancer Research (2015), 21 (10), 2278-2288CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: Infusion of interleukin-12 (IL12) can mediate antitumor immunity in animal models, yet its systemic administration to patients with cancer results in minimal efficacy and severe toxicity. Here, we evaluated the antitumor activity of adoptively transferred human tumor-infiltrating lymphocytes (TILs) genetically engineered to secrete single-chain IL12 selectively at the tumor site. Exptl. Design: Thirty-three patients with metastatic melanoma were treated in a cell dose-escalation trial of autologous TILs transduced with a gene encoding a single-chain IL12 driven by a nuclear factor of the activated T cells promoter (NFAT.IL12). No IL2 was administered. Results: The administration of 0.001 to 0.1 × 109 NFAT.IL12-transduced TILs to 17 patients resulted in a single, objective response (5.9%). However, at doses between 0.3 and 3 × 109 cells, 10 of 16 patients (63%) exhibited objective clin. responses. The responses tended to be short, and the administered IL12-producing cells rarely persisted at 1 mo. Increasing cell doses were assocd. with high serum levels of IL12 and IFNγ as well as clin. toxicities, including liver dysfunction, high fevers, and sporadic life-threatening hemodynamic instability. Conclusions: In this first-in-man trial, administration of TILs transduced with an inducible IL12 gene mediated tumor responses in the absence of IL2 administration using cell doses 10- to 100-fold lower than conventional TILs. However, due to toxicities, likely attributable to the secreted IL12, further refinement will be necessary before this approach can be safely used in the treatment of cancer patients.
From NLM Medline
- 53Pegram, H. J.; Lee, J. C.; Hayman, E. G.; Imperato, G. H.; Tedder, T. F.; Sadelain, M.; Brentjens, R. J. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood 2012, 119 (18), 4133– 4141, DOI: 10.1182/blood-2011-12-400044Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmvV2rsLg%253D&md5=c2c73f661b25d5c17d2f8a529328e4a3Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioningPegram, Hollie J.; Lee, James C.; Hayman, Erik G.; Imperato, Gavin H.; Tedder, Thomas F.; Sadelain, Michel; Brentjens, Renier J.Blood (2012), 119 (18), 4133-4141CODEN: BLOOAW; ISSN:0006-4971. (American Society of Hematology)Adoptive cell therapy with tumor-targeted T cells is a promising approach to cancer therapy. Enhanced clin. outcome using this approach requires conditioning regimens with total body irradn., lymphodepleting chemotherapy, and/or addnl. cytokine support. However, the need for prior conditioning precludes optimal application of this approach to a significant no. of cancer patients intolerant to these regimens. Herein, we present preclin. studies demonstrating that treatment with CD19-specific, chimeric antigen receptor (CAR)-modified T cells that are further modified to constitutively secrete IL-12 are able to safely eradicate established disease in the absence of prior conditioning. We demonstrate in a novel syngeneic tumor model that tumor elimination requires both CD4+ and CD8+ T-cell subsets, autocrine IL-12 stimulation, and subsequent IFNγ secretion by the CAR+ T cells. Importantly, IL-12-secreting, tumor-targeted T cells acquire intrinsic resistance to T regulatory cell-mediated inhibition. Based on these preclin. data, we anticipate that adoptive therapy using CAR-targeted T cells modified to secrete IL-12 will obviate or reduce the need for potentially hazardous conditioning regimens to achieve optimal antitumor responses in cancer patients.
From NLM Medline
- 54Gioux, S.; Choi, H. S.; Frangioni, J. V. Image-guided surgery using invisible near-infrared light: fundamentals of clinical translation. Mol. Imaging 2010, 9 (5), 237– 255, DOI: 10.2310/7290.2010.00034Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsVejtLbP&md5=08c56d952550d381f30e3f3f5f4e3b05Image-guided surgery using invisible near-infrared light: fundamentals of clinical translationGioux, Sylvain; Choi, Hak Soo; Frangioni, John V.Molecular Imaging (2010), 9 (5), 237-255CODEN: MIOMBP; ISSN:1535-3508. (BC Decker Inc.)A review. The field of biomedical optics has matured rapidly over the last decade and is poised to make a significant impact on patient care. In particular, wide-field (typically > 5 cm), planar, near-IR (NIR) fluorescence imaging has the potential to revolutionize human surgery by providing real-time image guidance to surgeons for tissue that needs to be resected, such as tumors, and tissue that needs to be avoided, such as blood vessels and nerves. However, to become a clin. reality, optimized imaging systems and NIR fluorescent contrast agents will be needed. In this review, we introduce the principles of NIR fluorescence imaging, analyze existing NIR fluorescence imaging systems, and discuss the key parameters that guide contrast agent development. We also introduce the complexities surrounding clin. translation using our experience with the Fluorescence-Assisted Resection and Exploration (FLARE) imaging system as an example. Finally, we introduce state-of-the-art optical imaging techniques that might someday improve image-guided surgery even further.
- 55Mondal, S. B.; Gao, S.; Zhu, N.; Liang, R.; Gruev, V.; Achilefu, S. Real-time fluorescence image-guided oncologic surgery. Adv. Cancer Res. 2014, 124, 171– 211, DOI: 10.1016/B978-0-12-411638-2.00005-7Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXptlOjsbs%253D&md5=5ab78702b3cd0c27e9e3062c583ece83Real-time fluorescence image-guided oncologic surgeryMondal, Suman B.; Gao, Shengkui; Zhu, Nan; Liang, Rongguang; Gruev, Viktor; Achilefu, SamuelAdvances in Cancer Research (2014), 124 (Emerging Applications of Molecular Imaging to Oncology), 171-211CODEN: ACRSAJ; ISSN:0065-230X. (Elsevier Inc.)Medical imaging plays a crit. role in cancer diagnosis and planning. Many of these patients rely on surgical intervention for curative outcomes. This requires a careful identification of the primary and microscopic tumors, and the complete removal of cancer. Although there have been efforts to adapt traditional-imaging modalities for intraoperative image guidance, they suffer from several constraints such as large hardware footprint, high-operation cost, and disruption of the surgical workflow. Because of the ease of image acquisition, relatively low-cost devices and intuitive operation, optical imaging methods have received tremendous interests for use in real-time image-guided surgery. To improve imaging depth under low interference by tissue autofluorescence, many of these applications utilize light in the near-IR (NIR) wavelengths, which is invisible to human eyes. With the availability of a wide selection of tumor-avid contrast agents, advancements in imaging sensors, electronic and optical designs, surgeons are able to combine different attributes of NIR optical imaging techniques to improve treatment outcomes. The emergence of diverse com. and exptl. image guidance systems, which are in various stages of clin. translation, attests to the potential high impact of intraoperative optical imaging methods to improve speed of oncol. surgery with high accuracy and minimal margin positivity.
- 56Stummer, W.; Pichlmeier, U.; Meinel, T.; Wiestler, O. D.; Zanella, F.; Reulen, H. J.; Group, A. L.-G. S. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 2006, 7 (5), 392– 401, DOI: 10.1016/S1470-2045(06)70665-9Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XjvFWlsb0%253D&md5=5f3656046a38c1f4324e4b7715d3995fFluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomized controlled multicenter phase III trialStummer, Walter; Pichlmeier, Uwe; Meinel, Thomas; Wiestler, Otmar Dieter; Zanella, Friedhelm; Reulen, Hans-Juergen; Oppel, F.; Brune, A.; Lanksch, W.; Woiciechowsky, C.; Brock, M.; Vesper, J.; Tonn, J-C.; Goetz, C.; Mayfrank, L.; Oertel, M. F.; Seifert, V.; Franz, K.; Bink, A.; Schackert, G.; Pinzer, T.; Hassler, W.; Bani, A.; Meisel, H-J.; Kern, B. C.; Mehdorn, H. M.; Nabavi, A.; Brawanski, A.; Ullrich, W. W.; Boeker, D. K.; Winking, M.; Weber, F.; Langenbach, U.; Kaehler, U.; Arnold, H.; Knopp, U.; Grumme, T.; Stretz, T.; Stolke, D.; Wiedemayer, H.; Turowski, B.; Pietsch, T.Lancet Oncology (2006), 7 (5), 392-401CODEN: LOANBN; ISSN:1470-2045. (Elsevier Ltd.)5-Aminolevulinic acid is a non-fluorescent prodrug that leads to intracellular accumulation of fluorescent porphyrins in malignant gliomas-a finding that is under investigation for intraoperative identification and resection of these tumors. We aimed to assess the effect of fluorescence-guided resection with 5-aminolevulinic acid on surgical radicality, progression-free survival, overall survival, and morbidity. Three hundred and twenty-two patients aged 23-73 years with suspected malignant glioma amenable to complete resection of contrast-enhancing tumor were randomly assigned to 20 mg/kg bodyweight 5-aminolevulinic acid for fluorescence-guided resection (n = 161) or to conventional microsurgery with white light (n = 161). The primary endpoints were the no. of patients without contrast-enhancing tumor on early MRI (ie, that obtained within 72 h after surgery) and 6-mo progression-free survival as assessed by MRI. Secondary endpoints were vol. of residual tumor on postoperative MRI, overall survival, neurol. deficit, and toxic effects. We report the results of an interim anal. with 270 patients in the full-anal. population (139 assigned 5-aminolevulinic acid, 131 assigned white light), which excluded patients with ineligible histol. and radiol. findings as assessed by central reviewers who were masked as to treatment allocation; the interim anal. resulted in termination of the study as defined by the protocol. Primary and secondary endpoints were analyzed by intention to treat in the full-anal. population. The study is registered at as NCT00241670. Median follow-up was 35.4 mo (95% CI 1.0-56.7). Contrast-enhancing tumor was reseted completely in 90 (65%) of 139 patients assigned 5-aminolevulinic acid compared with 47 (36%) of 131 assigned white light (difference between groups 29% [95% CI 17-40], p < 0.0001). Patients allocated 5-aminolevulinic acid had higher 6-mo progression free survival than did those allocated white light (41.0% [32.8-49.2] vs. 21.1% [14.0-28.2]; difference between groups 19.9% [9.1-30.7], p = 0.0003, Z test). Groups did not differ in the frequency of severe adverse events or adverse events in any organ system class reported within 7 days after surgery. Tumor fluorescence derived from 5-aminolevulinic acid enables more complete resection of contrast-enhancing tumor, leading to improved progression-free survival in patients with malignant glioma.
- 57Bardhan, A.; Deiters, A. Development of photolabile protecting groups and their application to the optochemical control of cell signaling. Curr. Opin. Struct. Biol. 2019, 57, 164– 175, DOI: 10.1016/j.sbi.2019.03.028Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXnvV2nurY%253D&md5=45fe067abf5f661ff7e572a31da408a8Development of photolabile protecting groups and their application to the optochemical control of cell signalingBardhan, Anirban; Deiters, AlexanderCurrent Opinion in Structural Biology (2019), 57 (), 164-175CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)Many biol. processes are naturally regulated with spatiotemporal control. In order to perturb and investigate them, optochem. tools have been developed that convey similar spatiotemporal precision. Pivotal to optochem. probes are photolabile protecting groups, so called caging groups, and recent developments have enabled new applications to cellular processes, including cell signaling. This review focuses on the advances made in the field of caging groups and their application in cell signaling through caged mols. such as neurotransmitters, lipids, secondary messengers, and proteins.
From NLM Medline
- 58Di Stasi, A.; Tey, S. K.; Dotti, G.; Fujita, Y.; Kennedy-Nasser, A.; Martinez, C.; Straathof, K.; Liu, E.; Durett, A. G.; Grilley, B. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 2011, 365 (18), 1673– 1683, DOI: 10.1056/NEJMoa1106152Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsVWqtbbF&md5=5adcc0e5acc341b06e4e9472959961f0Inducible apoptosis as a safety switch for adoptive cell therapyDi Stasi, Antonio; Tey, Siok-Keen; Dotti, Gianpietro; Fujita, Yuriko; Kennedy-Nasser, Alana; Martinez, Caridad; Straathof, Karin; Liu, Enli; Durett, April G.; Grilley, Bambi; Liu, Hao; Cruz, Conrad R.; Savoldo, Barbara; Gee, Adrian P.; Schindler, John; Krance, Robert A.; Heslop, Helen E.; Spencer, David M.; Rooney, Cliona M.; Brenner, Malcolm K.New England Journal of Medicine (2011), 365 (18), 1673-1683CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)A review. BACKGROUND Cellular therapies could play a role in cancer treatment and regenerative medicine if it were possible to quickly eliminate the infused cells in case of adverse events. We devised an inducible T-cell safety switch that is based on the fusion of human caspase 9 to a modified human FK-binding protein, allowing conditional dimerization. When exposed to a synthetic dimerizing drug, the inducible caspase 9 (iCasp9) becomes activated and leads to the rapid death of cells expressing this construct. METHODS We tested the activity of our safety switch by introducing the gene into donor T cells given to enhance immune reconstitution in recipients of haploidentical stem-cell transplants. Patients received AP1903, an otherwise bioinert small-mol. dimerizing drug, if graft-vs.-host disease (GVHD) developed. We measured the effects of AP1903 on GVHD and on the function and persistence of the cells contg. the iCasp9 safety switch. RESULTS Five patients between the ages of 3 and 17 years who had undergone stem-cell transplantation for relapsed acute leukemia were treated with the genetically modified T cells. The cells were detected in peripheral blood from all five patients and increased in no. over time, despite their constitutive transgene expression. A single dose of dimerizing drug, given to four patients in whom GVHD developed, eliminated more than 90% of the modified T cells within 30 min after administration and ended the GVHD without recurrence. CONCLUSIONS The iCasp9 cell-suicide system may increase the safety of cellular therapies and expand their clin. applications.
- 59Wu, C. Y.; Roybal, K. T.; Puchner, E. M.; Onuffer, J.; Lim, W. A. Remote control of therapeutic T cells through a small molecule-gated chimeric receptor. Science 2015, 350 (6258), aab4077, DOI: 10.1126/science.aab4077Google ScholarThere is no corresponding record for this reference.
- 60Reczek, C. R.; Chandel, N. S. The Two Faces of Reactive Oxygen Species in Cancer. Ann. Rev. Cancer Bio. 2017, 1 (1), 79– 98, DOI: 10.1146/annurev-cancerbio-041916-065808Google ScholarThere is no corresponding record for this reference.
- 61Sharma, A.; Arambula, J. F.; Koo, S.; Kumar, R.; Singh, H.; Sessler, J. L.; Kim, J. S. Hypoxia-targeted drug delivery. Chem. Soc. Rev. 2019, 48 (3), 771– 813, DOI: 10.1039/C8CS00304AGoogle Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFOms7nK&md5=37bbc627b91a138d5e8fdef25869dd29Hypoxia-targeted drug deliverySharma, Amit; Arambula, Jonathan F.; Koo, Seyoung; Kumar, Rajesh; Singh, Hardev; Sessler, Jonathan L.; Kim, Jong SeungChemical Society Reviews (2019), 48 (3), 771-813CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. Hypoxia is a state of low oxygen tension found in numerous solid tumors. It is typically assocd. with abnormal vasculature, which results in a reduced supply of oxygen and nutrients, as well as impaired delivery of drugs. The hypoxic nature of tumors often leads to the development of localized heterogeneous environments characterized by variable oxygen concns., relatively low pH, and increased levels of reactive oxygen species (ROS). The hypoxic heterogeneity promotes tumor invasiveness, metastasis, angiogenesis, and an increase in multidrug-resistant proteins. These factors decrease the therapeutic efficacy of anticancer drugs and can provide a barrier to advancing drug leads beyond the early stages of preclin. development. This review highlights various hypoxia-targeted and activated design strategies for the formulation of drugs or prodrugs and their mechanism of action for tumor diagnosis and treatment.
- 62Wang, M.; Zhao, J.; Zhang, L.; Wei, F.; Lian, Y.; Wu, Y.; Gong, Z.; Zhang, S.; Zhou, J.; Cao, K. Role of tumor microenvironment in tumorigenesis. J. Cancer 2017, 8 (5), 761– 773, DOI: 10.7150/jca.17648Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVCrsLnI&md5=1485e1cc6546c11fa3bcf6ab72c82b7bRole of tumor microenvironment in tumorigenesisWang, Maonan; Zhao, Jingzhou; Zhang, Lishen; Wei, Fang; Lian, Yu; Wu, Yingfeng; Gong, Zhaojian; Zhang, Shanshan; Zhou, Jianda; Cao, Ke; Li, Xiayu; Xiong, Wei; Li, Guiyuan; Zeng, Zhaoyang; Guo, CanJournal of Cancer (Wyoming, Australia) (2017), 8 (5), 761-773CODEN: JCWAAL; ISSN:1837-9664. (Ivyspring International Publisher Pty Ltd.)Tumorigenesis is a complex and dynamic process, consisting of three stages: initiation, progression, and metastasis. Tumors are encircled by extracellular matrix (ECM) and stromal cells, and the physiol. state of the tumor microenvironment (TME) is closely connected to every step of tumorigenesis. Evidence suggests that the vital components of the TME are fibroblasts and myofibroblasts, neuroendocrine cells, adipose cells, immune and inflammatory cells, the blood and lymphatic vascular networks, and ECM. This manuscript, based on the current studies of the TME, offers a more comprehensive overview of the primary functions of each component of the TME in cancer initiation, progression, and invasion. The manuscript also includes primary therapeutic targeting markers for each player, which may be helpful in treating tumors.
- 63Boedtkjer, E.; Pedersen, S. F. The Acidic Tumor Microenvironment as a Driver of Cancer. Annu. Rev. Physiol. 2020, 82 (1), 103– 126, DOI: 10.1146/annurev-physiol-021119-034627Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitFKitbjL&md5=72d46086784f0c569ef1d1086087af73The Acidic Tumor Microenvironment as a Driver of CancerBoedtkjer, Ebbe; Pedersen, Stine F.Annual Review of Physiology (2020), 82 (), 103-126CODEN: ARPHAD; ISSN:0066-4278. (Annual Reviews)A review. Acidic metabolic waste products accumulate in the tumor microenvironment because of high metabolic activity and insufficient perfusion. In tumors, the acidity of the interstitial space and the relatively well-maintained intracellular pH influence cancer and stromal cell function, their mutual interplay, and their interactions with the extracellular matrix. Tumor pH is spatially and temporally heterogeneous, and the fitness advantage of cancer cells adapted to extracellular acidity is likely particularly evident when they encounter less acidic tumor regions, for instance, during invasion. Through complex effects on genetic stability, epigenetics, cellular metab., proliferation, and survival, the compartmentalized pH microenvironment favors cancer development. Cellular selection exacerbates the malignant phenotype, which is further enhanced by acid-induced cell motility, extracellular matrix degrdn., attenuated immune responses, and modified cellular and intercellular signaling. In this review, we discuss how the acidity of the tumor microenvironment influences each stage in cancer development, from dysplasia to full-blown metastatic disease.
- 64Kochenderfer, J. N.; Feldman, S. A.; Zhao, Y.; Xu, H.; Black, M. A.; Morgan, R. A.; Wilson, W. H.; Rosenberg, S. A. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J. Immunother. 2009, 32 (7), 689– 702, DOI: 10.1097/CJI.0b013e3181ac6138Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXovFSisrs%253D&md5=422c0e7057c58dffbe5f4ab46e9f4b46Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptorKochenderfer, James N.; Feldman, Steven A.; Zhao, Yangbing; Xu, Hui; Black, Mary A.; Morgan, Richard A.; Wilson, Wyndham H.; Rosenberg, Steven A.Journal of Immunotherapy (2009), 32 (7), 689-702CODEN: JOIMF8; ISSN:1524-9557. (Lippincott Williams & Wilkins)T cells can be engineered to express the genes of chimeric antigen receptors (CARs) that recognize tumor-assocd. antigens. We constructed and compared 2 CARs that contained a single chain variable region moiety that recognized CD19. One CAR contained the signaling moiety of the 4-1BB mol. and the other did not. We selected the CAR that did not contain the 4-1BB moiety for further preclin. development. We demonstrated that gammaretroviruses encoding this receptor could transduce human T cells. Anti-CD19-CAR-transduced CD8 and CD4 T cells produced interferon-γ and interleukin-2 specifically in response to CD19 target cells. The transduced T cells specifically killed primary chronic lymphocytic leukemia (CLL) cells. We transduced T cells from CLL patients that had been previously treated with chemotherapy. We induced these T cells to proliferate sufficiently to provide enough cells for clin. adoptive T cell transfer with a protocol consisting of an initial stimulation with an anti-CD3 monoclonal antibody (OKT3) before transduction followed by a second OKT3 stimulation 7 days after transduction. This protocol was successfully adapted for use in CLL patients with high peripheral blood leukemia cell counts by depleting CD19 cells before the initial OKT3 stimulation. In prepn. for a clin. trial that will enroll patients with advanced B cell malignancies, we generated a producer cell clone that produces retroviruses encoding the anti-CD19 CAR, and we produced sufficient retroviral supernatant for the proposed clin. trial under good manufg. practice conditions.
From NLM Medline
Cited By
This article is cited by 1 publications.
- Alison C. Leonard, Anika J. Friedman, Rachel Chayer, Brian M. Petersen, Janty Woojuh, Zenan Xing, Sean R. Cutler, Joel L. Kaar, Michael R. Shirts, Timothy A. Whitehead. Rationalizing Diverse Binding Mechanisms to the Same Protein Fold: Insights for Ligand Recognition and Biosensor Design. ACS Chemical Biology 2024, 19
(8)
, 1757-1772. https://doi.org/10.1021/acschembio.4c00243
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 64 other publications.
- 1Gross, G.; Waks, T.; Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl. Acad. Sci. U.S.A. 1989, 86 (24), 10024– 10028, DOI: 10.1073/pnas.86.24.100241https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXntFChsw%253D%253D&md5=3be7a08d7d83af1c25a740bdc0ab670fExpression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificityGross, Gideon; Waks, Tova; Eshhar, ZeligProceedings of the National Academy of Sciences of the United States of America (1989), 86 (24), 10024-8CODEN: PNASA6; ISSN:0027-8424.To design and direct at will the specificity of T cells in a non-major histocompatibility complex (MHC)-restricted manner, chimeric T-cell receptor (TcR) genes composed of the TcR const. (C) domains fused to the antibody's variable (V) domains were constructed and expressed. Genomic expression vectors were constructed contg. the rearranged gene segments coding for the V region domains of the heavy (VH) and light (VL) chains of an anti-2,4,6-trinitrophenyl (TNP) antibody (SP6) spliced to either one of the C-region gene segments of the α or β TcR chains. Following transfection into a cytotoxic T-cell hybridoma, expression of a functional TcR was detected. The chimeric TcR exhibited the idiotope of the Sp6 anti-TNP antibody and endowed the T cells with a non-MHC-restricted response to the hapten TNP. The transfectants specifically killed and produced interleukin 2 in response to TNP-bearing target cells across strain and species barriers. Moreover, such transfectants responded to immobilized TNP-protein conjugates, bypassing the need for cellular processing and presentation. In the particular system employed, both the TNP-binding site and the Sp6 idiotope reside almost exclusively in the VH chain region. Hence, introduction into T cells of TcR genes contg. only the VHSp6 fused to either the Cα or Cβ was sufficient for the expression of a functional surface receptor. Apparently, the VHCα or VHCβ chimeric chains can pair with the endogenous β or α chains of the recipient T cell to form a functional αβ heterodimeric receptor. Thus, this chimeric receptor provides the T cell with an antibody-like specificity and is able to effectively transmit the signal for T-cell activation and execution of its effector function.
- 2Sadelain, M.; Brentjens, R.; Riviere, I. The basic principles of chimeric antigen receptor design. Cancer Discovery 2013, 3 (4), 388– 398, DOI: 10.1158/2159-8290.CD-12-05482https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXlvVCit7c%253D&md5=ebd63df454ec54c889f874c1dc59d669The Basic Principles of Chimeric Antigen Receptor DesignSadelain, Michel; Brentjens, Renier; Riviere, IsabelleCancer Discovery (2013), 3 (4), 388-398CODEN: CDAIB2; ISSN:2159-8274. (American Association for Cancer Research)A review. Chimeric antigen receptors (CAR) are recombinant receptors that provide both antigen-binding and T-cell-activating functions. A multitude of CARs has been reported over the past decade, targeting an array of cell surface tumor antigens. Their biol. functions have dramatically changed following the introduction of tripartite receptors comprising a costimulatory domain, termed second-generation CARs. These have recently shown clin. benefit in patients treated with CD19-targeted autologous T cells. CARs may be combined with costimulatory ligands, chimeric costimulatory receptors, or cytokines to further enhance T-cell potency, specificity, and safety. CARs represent a new class of drugs with exciting potential for cancer immunotherapy. Significance: CARs are a new class of drugs with great potential for cancer immunotherapy. Upon their expression in T lymphocytes, CARs direct potent, targeted immune responses that have recently shown encouraging clin. outcomes in a subset of patients with B-cell malignancies. This review focuses on the design of CARs, including the requirements for optimal antigen recognition and different modalities to provide costimulatory support to targeted T cells, which include the use of second- and third-generation CARs, costimulatory ligands, chimeric costimulatory receptors, and cytokines. Cancer Discov; 3(4); 388-98. ©2013 AACR.
- 3June, C. H.; Sadelain, M. Chimeric Antigen Receptor Therapy. N. Engl. J. Med. 2018, 379 (1), 64– 73, DOI: 10.1056/NEJMra17061693https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlert7rL&md5=87cbf53139fed4142b1529e6f25f0ea5Chimeric antigen receptor therapyJune, Carl H.; Sadelain, MichelNew England Journal of Medicine (2018), 379 (1), 64-73CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)The present article describes about principles of T-cell engineering and synthetic immunity, with focus on efficacy and toxic effects of CAR therapies.
- 4Lim, W. A.; June, C. H. The Principles of Engineering Immune Cells to Treat Cancer. Cell 2017, 168 (4), 724– 740, DOI: 10.1016/j.cell.2017.01.0164https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXis1ygtLs%253D&md5=2933e55842cae207c5258ca3934eeb2eThe principles of engineering immune cells to treat cancerLim, Wendell A.; June, Carl H.Cell (Cambridge, MA, United States) (2017), 168 (4), 724-740CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. Chimeric antigen receptor (CAR) T cells have proven that engineered immune cells can serve as a powerful new class of cancer therapeutics. Clin. experience has helped to define the major challenges that must be met to make engineered T cells a reliable, safe, and effective platform that can be deployed against a broad range of tumors. The emergence of synthetic biol. approaches for cellular engineering is providing us with a broadly expanded set of tools for programming immune cells. We discuss how these tools could be used to design the next generation of smart T cell precision therapeutics.
- 5Urbanska, K.; Lanitis, E.; Poussin, M.; Lynn, R. C.; Gavin, B. P.; Kelderman, S.; Yu, J.; Scholler, N.; Powell, D. J. A Universal Strategy for Adoptive Immunotherapy of Cancer through Use of a Novel T-cell Antigen Receptor. Cancer Res. 2012, 72 (7), 1844– 1852, DOI: 10.1158/0008-5472.CAN-11-38905https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XkvVeltrs%253D&md5=b46e16be9bee35426d0a9ff2108bbb6dA Universal Strategy for Adoptive Immunotherapy of Cancer through Use of a Novel T-cell Antigen ReceptorUrbanska, Katarzyna; Lanitis, Evripidis; Poussin, Mathilde; Lynn, Rachel C.; Gavin, Brian P.; Kelderman, Sander; Yu, Jason; Scholler, Nathalie; Powell, Daniel J., Jr.Cancer Research (2012), 72 (7), 1844-1852CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Adoptive immunotherapies composed of T cells engineered to express a chimeric antigen receptor (CAR) offer an attractive strategy for treatment of human cancer. However, CARs have a fixed antigen specificity such that only one tumor-assocd. antigen (TAA) can be targeted, limiting the efficacy that can be achieved because of heterogeneous TAA expression. For this reason, a more generalized and effective application of CAR therapy would benefit from the capability to produce large panels of CARs against many known TAAs. In this study, we show a novel strategy to extend the recognition specificity potential of a bioengineered lymphocyte population, allowing flexible approaches to redirect T cells against various TAAs. Our strategy employs a biotin-binding immune receptor (BBIR) composed of an extracellular-modified avidin linked to an intracellular T-cell signaling domain. BBIR T cells recognized and bound exclusively to cancer cells pretargeted with specific biotinylated mols. The versatility afforded by BBIRs permitted sequential or simultaneous targeting of a combination of distinct antigens. Together, our findings show that a platform of universal T-cell specificity can significantly extend conventional CAR approaches, permitting the tailored generation of T cells of unlimited antigen specificity for improving the effectiveness of adoptive T-cell immunotherapies for cancer. Cancer Res; 72(7); 1844-52.
- 6Lee, D. W.; Kochenderfer, J. N.; Stetler-Stevenson, M.; Cui, Y. K.; Delbrook, C.; Feldman, S. A.; Fry, T. J.; Orentas, R.; Sabatino, M.; Shah, N. N. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 2015, 385 (9967), 517– 528, DOI: 10.1016/S0140-6736(14)61403-36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhslGjtLbM&md5=6d2ae3cb6d1672a1f0b3c0fedef2c337T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trialLee, Daniel W.; Kochenderfer, James N.; Stetler-Stevenson, Maryalice; Cui, Yongzhi K.; Delbrook, Cindy; Feldman, Steven A.; Fry, Terry J.; Orentas, Rimas; Sabatino, Marianna; Shah, Nirali N.; Steinberg, Seth M.; Stroncek, Dave; Tschernia, Nick; Yuan, Constance; Zhang, Hua; Zhang, Ling; Rosenberg, Steven A.; Wayne, Alan S.; Mackall, Crystal L.Lancet (2015), 385 (9967), 517-528CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)Chimeric antigen receptor (CAR) modified T cells targeting CD19 have shown activity in case series of patients with acute and chronic lymphocytic leukemia and B-cell lymphomas, but feasibility, toxicity, and response rates of consecutively enrolled patients treated with a consistent regimen and assessed on an intention-to-treat basis have not been reported. We aimed to define feasibility, toxicity, max. tolerated dose, response rate, and biol. correlates of response in children and young adults with refractory B-cell malignancies treated with CD19-CAR T cells. This phase 1, dose-escalation trial consecutively enrolled children and young adults (aged 1-30 years) with relapsed or refractory acute lymphoblastic leukemia or non-Hodgkin lymphoma. Autologous T cells were engineered via an 11-day manufg. process to express a CD19-CAR incorporating an anti-CD19 single-chain variable fragment plus TCR zeta and CD28 signalling domains. All patients received fludarabine and cyclophosphamide before a single infusion of CD19-CAR T cells. Using a std. 3 + 3 design to establish the max. tolerated dose, patients received either 1 × 106 CAR-transduced T cells per kg (dose 1), 3 × 106 CAR-transduced T cells per kg (dose 2), or the entire CAR T-cell product if sufficient nos. of cells to meet the assigned dose were not generated. After the dose-escalation phase, an expansion cohort was treated at the max. tolerated dose. The trial is registered with ClinicalTrials.gov, no. NCT01593696. Between July 2, 2012, and June 20, 2014, 21 patients (including eight who had previously undergone allogeneic haematopoietic stem-cell transplantation) were enrolled and infused with CD19-CAR T cells. 19 received the prescribed dose of CD19-CAR T cells, whereas the assigned dose concn. could not be generated for two patients (90% feasible). All patients enrolled were assessed for response. The max. tolerated dose was defined as 1 × 106 CD19-CAR T cells per kg. All toxicities were fully reversible, with the most severe being grade 4 cytokine release syndrome that occurred in three (14%) of 21 patients (95% CI 3·0-36·3). The most common non-haematol. grade 3 adverse events were fever (nine [43%] of 21 patients), hypokalemia (nine [43%] of 21 patients), fever and neutropenia (eight [38%] of 21 patients), and cytokine release syndrome (three [14%) of 21 patients). CD19-CAR T cell therapy is feasible, safe, and mediates potent anti-leukemic activity in children and young adults with chemotherapy-resistant B-precursor acute lymphoblastic leukemia. All toxicities were reversible and prolonged B-cell aplasia did not occur. National Institutes of Health Intramural funds and St Baldrick's Foundation.
- 7Maude, S. L.; Frey, N.; Shaw, P. A.; Aplenc, R.; Barrett, D. M.; Bunin, N. J.; Chew, A.; Gonzalez, V. E.; Zheng, Z.; Lacey, S. F. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014, 371 (16), 1507– 1517, DOI: 10.1056/NEJMoa14072227https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVSls73K&md5=7f13aaa25ae118901d3bec614b07cc3aChimeric antigen receptor T cells for sustained remissions in leukemiaMaude, Shannon L.; Frey, Noelle; Shaw, Pamela A.; Aplenc, Richard; Barrett, David M.; Bunin, Nancy J.; Chew, Anne; Gonzalez, Vanessa E.; Zheng, Zhaohui; Lacey, Simon F.; Mahnke, Yolanda D.; Melenhorst, Jan J.; Rheingold, Susan R.; Shen, Angela; Teachey, David T.; Levine, Bruce L.; June, Carl H.; Porter, David L.; Grupp, Stephan A.New England Journal of Medicine (2014), 371 (16), 1507-1517, 11 pp.CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)Background: Relapsed acute lymphoblastic leukemia (ALL) is difficult to treat despite the availability of aggressive therapies. Chimeric antigen receptor-modified T cells targeting CD19 may overcome many limitations of conventional therapies and induce remission in patients with refractory disease. Methods: We infused autologous T cells transduced with a CD19-directed chimeric antigen receptor (CTL019) lentiviral vector in patients with relapsed or refractory ALL at doses of 0.76 × 106 to 20.6 × 106 CTL019 cells per kg of body wt. Patients were monitored for a response, toxic effects, and the expansion and persistence of circulating CTL019 T cells. Results: A total of 30 children and adults received CTL019. Complete remission was achieved in 27 patients (90%), including 2 patients with blinatumomab-refractory disease and 15 who had undergone stem-cell transplantation. CTL019 cells proliferated in vivo and were detectable in the blood, bone marrow, and cerebrospinal fluid of patients who had a response. Sustained remission was achieved with a 6-mo event-free survival rate of 67% (95% confidence interval [CI], 51 to 88) and an overall survival rate of 78% (95% CI, 65 to 95). At 6 mo, the probability that a patient would have persistence of CTL019 was 68% (95% CI, 50 to 92) and the probability that a patient would have relapse-free B-cell aplasia was 73% (95% CI, 57 to 94). All the patients had the cytokine-release syndrome. Severe cytokine-release syndrome, which developed in 27% of the patients, was assocd. with a higher disease burden before infusion and was effectively treated with the anti-interleukin-6 receptor antibody tocilizumab. Conclusions: Chimeric antigen receptor-modified T-cell therapy against CD19 was effective in treating relapsed and refractory ALL. CTL019 was assocd. with a high remission rate, even among patients for whom stem-cell transplantation had failed, and durable remissions up to 24 mo were obsd.
- 8Park, J. H.; Riviere, I.; Gonen, M.; Wang, X.; Senechal, B.; Curran, K. J.; Sauter, C.; Wang, Y.; Santomasso, B.; Mead, E. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378 (5), 449– 459, DOI: 10.1056/NEJMoa17099198https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlylsbY%253D&md5=a2950f5baba38e254f35f812c56a80e9Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemiaPark, Jae H.; Riviere, Isabelle; Gonen, Mithat; Wang, Xiuyan; Senechal, Brigitte; Curran, Kevin J.; Sauter, Craig; Wang, Yongzeng; Santomasso, Bianca; Mead, Elena; Roshal, Mikhail; Maslak, Peter; Davila, Marco; Brentjens, Renier J.; Sadelain, MichelNew England Journal of Medicine (2018), 378 (5), 449-459CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)CD19-specific chimeric antigen receptor (CAR) T cells induce high rates of initial response among patients with relapsed B-cell acute lymphoblastic leukemia (ALL) and long-term remissions in a subgroup of patients. We conducted a phase 1 trial involving adults with relapsed B-cell ALL who received an infusion of autologous T cells expressing the 19-28z CAR at the Memorial Sloan Kettering Cancer Center (MSKCC). Safety and long-term outcomes were assessed, as were their assocns. with demog., clin., and disease characteristics. A total of 53 adults received 19-28z CAR T cells that were manufd. at MSKCC. After infusion, severe cytokine release syndrome occurred in 14 of 53 patients (26%; 95% confidence interval [CI], 15 to 40); 1 patient died. Complete remission was obsd. in 83% of the patients. At a median follow-up of 29 mo (range, 1 to 65), the median event-free survival was 6.1 mo (95% CI, 5.0 to 11.5), and the median overall survival was 12.9 mo (95% CI, 8.7 to 23.4). Patients with a low disease burden (<5% bone marrow blasts) before treatment had markedly enhanced remission duration and survival, with a median event-free survival of 10.6 mo (95% CI, 5.9 to not reached) and a median overall survival of 20.1 mo (95% CI, 8.7 to not reached). Patients with a higher burden of disease (≥5% bone marrow blasts or extramedullary disease) had a greater incidence of the cytokine release syndrome and neurotoxic events and shorter long-term survival than did patients with a low disease burden. In the entire cohort, the median overall survival was 12.9 mo. Among patients with a low disease burden, the median overall survival was 20.1 mo and was accompanied by a markedly lower incidence of the cytokine release syndrome and neurotoxic events after 19-28z CAR T-cell infusion than was obsd. among patients with a higher disease burden.
- 9Munshi, N. C.; Anderson, L. D., Jr; Shah, N.; Madduri, D.; Berdeja, J.; Lonial, S.; Raje, N.; Lin, Y.; Siegel, D.; Oriol, A. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N. Engl. J. Med. 2021, 384 (8), 705– 716, DOI: 10.1056/NEJMoa20248509https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXlt1KgsLY%253D&md5=730eaa169adca5221b86ece1884c18f3Idecabtagene vicleucel in relapsed and refractory multiple myelomaMunshi, Nikhil C.; Anderson, Larry D., Jr.; Shah, Nina; Madduri, Deepu; Berdeja, Jesus; Lonial, Sagar; Raje, Noopur; Lin, Yi; Siegel, David; Oriol, Albert; Moreau, Philippe; Yakoub-Agha, Ibrahim; Delforge, Michel; Cavo, Michele; Einsele, Hermann; Goldschmidt, Hartmut; Weisel, Katja; Rambaldi, Alessandro; Reece, Donna; Petrocca, Fabio; Massaro, Monica; Connarn, Jamie N.; Kaiser, Shari; Patel, Payal; Huang, Liping; Campbell, Timothy B.; Hege, Kristen; San-Miguel, JesusNew England Journal of Medicine (2021), 384 (8), 705-716CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)Idecabtagene vicleucel (ide-cel, also called bb2121), a B-cell maturation antigen-directed chimeric antigen receptor (CAR) T-cell therapy, has shown clin. activity with expected CAR T-cell toxic effects in patients with relapsed and refractory multiple myeloma. methods In this phase 2 study, we sought to confirm the efficacy and safety of ide-cel in patients with relapsed and refractory myeloma. Patients with disease after at least three previous regimens including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 antibody were enrolled. Patients received ide-cel target doses of 150 x 106 to 450 x 106 CAR-pos. (CAR+) T cells. The primary end point was an overall response (partial response or better); a key secondary end point was a complete response or better (comprising complete and stringent complete responses). results Of 140 patients enrolled, 128 received ide-cel. At a median follow-up of 13.3 mo, 94 of 128 patients (73%) had a response, and 42 of 128 (33%) had a complete response or better. Minimal residual disease (MRD)-neg. status (<10-5 nucleated cells) was confirmed in 33 patients, representing 26% of all 128 patients who were treated and 79% of the 42 patients who had a complete response or better. The median progression-free survival was 8.8 mo (95% confidence interval, 5.6 to 11.6). Common toxic effects among the 128 treated patients included neutropenia in 117 patients (91%), anemia in 89 (70%), and thrombocytopenia in 81 (63%). Cytokine release syndrome was reported in 107 patients (84%), including 7 (5%) who had events of grade 3 or higher. Neurotoxic effects developed in 23 patients (18%) and were of grade 3 in 4 patients (3%); no neurotoxic effects higher than grade 3 occurred. Cellular kinetic anal. confirmed CAR+ T cells in 29 of 49 patients (59%) at 6 mo and 4 of 11 patients (36%) at 12 mo after infusion. conclusions Ide-cel induced responses in a majority of heavily pretreated patients with refractory and relapsed myeloma; MRD-neg. status was achieved in 26% of treated patients. Almost all patients had grade 3 or 4 toxic effects, most commonly hematol. toxic effects and cytokine release syndrome.
From NLM Medline
- 10Ramakrishna, S.; Barsan, V.; Mackall, C. Prospects and challenges for use of CAR T cell therapies in solid tumors. Expert Opin. Biol. Ther. 2020, 20 (5), 503– 516, DOI: 10.1080/14712598.2020.173837810https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvVertLo%253D&md5=d9328ebe64880ed45af37bd1b84aa7cbProspects and challenges for use of CAR T cell therapies in solid tumorsRamakrishna, Sneha; Barsan, Valentin; Mackall, CrystalExpert Opinion on Biological Therapy (2020), 20 (5), 503-516CODEN: EOBTA2; ISSN:1471-2598. (Taylor & Francis Ltd.)A review. : Chimeric antigen receptor (CAR) T cell therapy has provided patients with relapsed/refractory B cell malignancies a new therapeutic option, but this class of therapeutics has not demonstrated consistent therapeutic benefit in solid tumors.: Here we review the literature to identify numerous factors that contribute to this discrepancy, using pediatric cancers as a platform to understand these limitations. We discuss an inability to target highly and homogenously expressed lineage-assocd. antigens due to risks of on-target off-tumor toxicity, T cell dysfunction related to T cell exhaustion and the suppressive tumor microenvironment (TME), and inefficient CAR T cell trafficking into solid tumors. As our understanding of the biol. of CAR T cells improves and innovations in engineering CAR platforms emerge, next generation CAR T cell therapeutics designed to overcome these challenges will enter the clinic for testing.: New approaches to address the challenges that have limited the efficacy of CAR T cell therapeutics in solid tumors are emerging. These approaches include next-generation CAR T cell engineering to overcome antigen heterogeneity, to mitigate T cell exhaustion and to prevent suppression by the TME, as well as novel approaches for regional delivery to facilitate tumor T cell trafficking.
- 11Maldini, C. R.; Ellis, G. I.; Riley, J. L. CAR T cells for infection, autoimmunity and allotransplantation. Nat. Rev. Immunol. 2018, 18 (10), 605– 616, DOI: 10.1038/s41577-018-0042-211https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlyrurbF&md5=ab83fb3b4a2abac6b439fbc5e560ac90CAR T cells for infection, autoimmunity and allotransplantationMaldini, Colby R.; Ellis, Gavin I.; Riley, James L.Nature Reviews Immunology (2018), 18 (10), 605-616CODEN: NRIABX; ISSN:1474-1733. (Nature Research)Chimeric antigen receptors (CARs) have shown remarkable ability to re-direct T cells to target CD19-expressing tumors, resulting in remission rates of up to 90% in individuals with paediatric acute lymphoblastic lymphoma. Lessons learned from these clin. trials of adoptive T cell therapy for cancer, as well as investments made in manufg. T cells at com. scale, have inspired researchers to develop CARs for addnl. applications. Here, we explore the challenges and opportunities of using this technol. to target infectious diseases such as with HIV and undesired immune responses such as autoimmunity and transplant rejection. Despite substantial obstacles, the potential of CAR T cells to enable cures for a wide array of disease settings could be transformational for the medical field.
- 12Bertoletti, A.; Tan, A. T. Challenges of CAR- and TCR-T cell-based therapy for chronic infections. J. Exp. Med. 2020, 217 (5), e20191663 DOI: 10.1084/jem.2019166312https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXovFSntrs%253D&md5=86d61cc45857a869f78b0525abefe18dChallenges of CAR- and TCR-T cell-based therapy for chronic infectionsBertoletti, Antonio; Tan, Anthony TanotoJournal of Experimental Medicine (2020), 217 (5), e20191663CODEN: JEMEAV; ISSN:1540-9538. (Rockefeller University Press)A review. While therapy with T cells engineered with a chimeric antigen receptor (CAR) or a classical T cell receptor (TCR) is revolutionizing cancer treatment, its adoption in infectious diseases has been met with considerable resistance. Can we find its value for the cure of infections.
- 13Press, M. F.; Cordon-Cardo, C.; Slamon, D. J. Expression of the HER-2/neu proto-oncogene in normal human adult and fetal tissues. Oncogene 1990, 5 (7), 953– 96213https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXkvFalsbc%253D&md5=1c839969b674279e8820f7575c2ecae7Expression of the HER-2/neu proto-oncogene in normal human adult and fetal tissuesPress, Michael F.; Cordon-Cardo, Carlos; Slamon, Dennis J.Oncogene (1990), 5 (7), 953-62CODEN: ONCNES; ISSN:0950-9232.The HER-2/neu proto-oncogene is homologous with, but distinct from the epidermal growth factor receptor. A variety of normal adult and fetal tissues were evaluated for HER-2/neu expression using immunohistochem. and Northern blot analyses to identify the HER-2/neu protein and transcript resp.. HER-2/neu protein was identified on cell membranes of epithelial cells in the gastrointestinal, respiratory, reproductive, and urinary tract as well as in the skin, breast and placenta. Northern hybridization confirmed the presence of the 4.5 kb transcript encoding the protein in these tissues. The amt. of HER-2/neu message and protein was generally higher in fetal tissues than in the corresponding normal adult tissues. HER-2/neu expression levels in these normal tissues were similar to the levels found in non-amplified, non-overexpresing breast cancers and breast cancer cell lines. Southern hybridization of extd. DNA showed that none of the normal tissues expressing HER-2/neu had amplification of the gene. These results confirm that HER-2/neu is normally a membrane constituent of a variety of epithelial cell types.
- 14Yano, S.; Kondo, K.; Yamaguchi, M.; Richmond, G.; Hutchison, M.; Wakeling, A.; Averbuch, S.; Wadsworth, P. Distribution and function of EGFR in human tissue and the effect of EGFR tyrosine kinase inhibition. Anticancer Res. 2003, 23 (5A), 3639– 365014https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXkvFWn&md5=70a289e6c8c5492e9c3537d77880af8dDistribution and function of EGFR in human tissue and the effect of EGFR tyrosine kinase inhibitionYano, Seiichi; Kondo, Kaoru; Yamaguchi, Motonori; Richmond, Graham; Hutchison, Michael; Wakeling, Alan; Averbuch, Steven; Wadsworth, PeterAnticancer Research (2003), 23 (5A), 3639-3650CODEN: ANTRD4; ISSN:0250-7005. (International Institute of Anticancer Research)A review. From immunohistochem. and ligand-binding studies, it is known that the epidermal growth factor receptor (EGFR), a member of the erbB family of receptors, is expressed in tissues of epithelial, mesenchymal and neuronal origin and plays a major role in normal cellular processes such as proliferation, differentiation and development. EGFR is highly expressed in a no. of solid tumors and its expression correlates with tumor progression, resistance to chemotherapy and a poor prognosis; it is consequently an attractive target for the rational design of novel anticancer agents. Knowledge of the role of EGFR in normal tissues will help the understanding of the adverse events assocd. with such agents. Studies in knockout mice and preclin. toxicol. studies have shown that the major effects of inhibiting the EGFR are skin and, gastrointestinal toxicities. Clin. studies with inhibitors of EGFR, such as gefitinib, cetuximab and erlotinib, have shown a favorable adverse-event profile, primarily consisting of skin and gastrointestinal toxicities, as predicted from the mechanism-based effects obsd. in preclin. studies.
- 15Majzner, R. G.; Mackall, C. L. Clinical lessons learned from the first leg of the CAR T cell journey. Nat. Med. 2019, 25 (9), 1341– 1355, DOI: 10.1038/s41591-019-0564-615https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslelurbN&md5=ab243f9a176dc0e3f9d65aebb9097021Clinical lessons learned from the first leg of the CAR T cell journeyMajzner, Robbie G.; Mackall, Crystal L.Nature Medicine (New York, NY, United States) (2019), 25 (9), 1341-1355CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)Chimeric antigen receptor (CAR) T cell therapy for B cell malignancies has surpassed expectations, driving an ever-expanding no. of clin. trials and the first US Food and Drug Administration approvals of cell therapies for the treatment of cancer. This experience has illuminated some generalizable requirements for CAR T cell efficacy as well as the interplay between disease biol. and clin. outcomes. Major CAR intrinsic variables affecting T cell behavior have been defined, and mechanisms of tumor resistance are increasingly understood. Here, we review the clin. experience with CAR T cells amassed to date, including but not limited to B cell malignancies, emphasizing factors assocd. with efficacy, resistance and major barriers to success. We also discuss how these insights are driving next-generation clin. trials, including those in solid tumors.
- 16Kershaw, M. H.; Westwood, J. A.; Parker, L. L.; Wang, G.; Eshhar, Z.; Mavroukakis, S. A.; White, D. E.; Wunderlich, J. R.; Canevari, S.; Rogers-Freezer, L. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin. Cancer Res. 2006, 12 (20), 6106– 6115, DOI: 10.1158/1078-0432.CCR-06-118316https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFWhsbnL&md5=a6a439a0187f50d8a22e12dfc1da89e5A Phase I Study on Adoptive Immunotherapy Using Gene-Modified T Cells for Ovarian CancerKershaw, Michael H.; Westwood, Jennifer A.; Parker, Linda L.; Wang, Gang; Eshhar, Zelig; Mavroukakis, Sharon A.; White, Donald E.; Wunderlich, John R.; Canevari, Silvana; Rogers-Freezer, Linda; Chen, Clara C.; Yang, James C.; Rosenberg, Steven A.; Hwu, PatrickClinical Cancer Research (2006), 12 (20, Pt. 1), 6106-6115CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: A phase I study was conducted to assess the safety of adoptive immunotherapy using gene-modified autologous T cells for the treatment of metastatic ovarian cancer. Exptl. Design: T cells with reactivity against the ovarian cancer-assocd. antigen α-folate receptor (FR) were generated by genetic modification of autologous T cells with a chimeric gene incorporating an anti-FR single-chain antibody linked to the signaling domain of the Fc receptor γ chain. Patients were assigned to one of two cohorts in the study. Eight patients in cohort 1 received a dose escalation of T cells in combination with high-dose interleukin-2, and six patients in cohort 2 received dual-specific T cells (reactive with both FR and allogeneic cells) followed by immunization with allogeneic peripheral blood mononuclear cells. Results: Five patients in cohort 1 experienced some grade 3 to 4 treatment-related toxicity that was probably due to interleukin-2 administration, which could be managed using std. measures. Patients in cohort 2 experienced relatively mild side effects with grade 1 to 2 symptoms. No redn. in tumor burden was seen in any patient. Tracking 111In-labeled adoptively transferred T cells in cohort 1 revealed a lack of specific localization of T cells to tumor except in one patient where some signal was detected in a peritoneal deposit. PCR anal. showed that gene-modified T cells were present in the circulation in large nos. for the first 2 days after transfer, but these quickly declined to be barely detectable 1 mo later in most patients. An inhibitory factor developed in the serum of three of six patients tested over the period of treatment, which significantly reduced the ability of gene-modified T cells to respond against FR+ tumor cells. Conclusions: Large nos. of gene-modified tumor-reactive T cells can be safely given to patients, but these cells do not persist in large nos. long term. Future studies need to employ strategies to extend T cell persistence. This report is the first to document the use of genetically redirected T cells for the treatment of ovarian cancer.
- 17Lamers, C. H.; Sleijfer, S.; Vulto, A. G.; Kruit, W. H.; Kliffen, M.; Debets, R.; Gratama, J. W.; Stoter, G.; Oosterwijk, E. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J. Clin. Oncol. 2006, 24 (13), e20– e22, DOI: 10.1200/JCO.2006.05.9964There is no corresponding record for this reference.
- 18Maus, M. V.; Haas, A. R.; Beatty, G. L.; Albelda, S. M.; Levine, B. L.; Liu, X.; Zhao, Y.; Kalos, M.; June, C. H. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol. Res. 2013, 1 (1), 26– 31, DOI: 10.1158/2326-6066.CIR-13-000618https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtFSjtLo%253D&md5=39a36c1c1c297760ad7f7298c09df899T cells expressing chimeric antigen receptors can cause anaphylaxis in humansMaus, Marcela V.; Haas, Andrew R.; Beatty, Gregory L.; Albelda, Steven M.; Levine, Bruce L.; Liu, Xiaojun; Zhao, Yangbing; Kalos, Michael; June, Carl H.Cancer Immunology Research (2013), 1 (1), 26-31CODEN: CIRACV; ISSN:2326-6066. (American Association for Cancer Research)T cells can be redirected to overcome tolerance to cancer by engineering with integrating vectors to express a chimeric antigen receptor (CAR). In preclin. models, we have previously shown that transfection of T cells with mRNA coding for a CAR is an alternative strategy that has antitumor efficacy and the potential to evaluate the on-target off-tumor toxicity of new CAR targets safely due to transient mRNA CAR expression. Here, we report the safety obsd. in four patients treated with autologous T cells that had been electroporated with mRNA coding for a CAR derived from a murine antibody to human mesothelin. Because of the transient nature of CAR expression on the T cells, subjects in the clin. study were given repeated infusions of the CAR-T cells to assess their safety. One subject developed anaphylaxis and cardiac arrest within minutes of completing the third infusion. Although human anti-mouse Ig (Ig)G antibodies have been known to develop with CAR-transduced T cells, they have been thought to have no adverse clin. consequences. This is the first description of clin. anaphylaxis resulting from CAR-modified T cells, most likely through IgE antibodies specific to the CAR. These results indicate that the potential immunogenicity of CARs derived from murine antibodies may be a safety issue for mRNA CARs, esp. when administered using an intermittent dosing schedule.
- 19Roybal, K. T.; Rupp, L. J.; Morsut, L.; Walker, W. J.; McNally, K. A.; Park, J. S.; Lim, W. A. Precision Tumor Recognition by T Cells With Combinatorial Antigen-Sensing Circuits. Cell 2016, 164 (4), 770– 779, DOI: 10.1016/j.cell.2016.01.01119https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xhs12gt78%253D&md5=dc3da3e426700a1dd4f57417155c97b5Precision Tumor Recognition by T Cells With Combinatorial Antigen-Sensing CircuitsRoybal, Kole T.; Rupp, Levi J.; Morsut, Leonardo; Walker, Whitney J.; McNally, Krista A.; Park, Jason S.; Lim, Wendell A.Cell (Cambridge, MA, United States) (2016), 164 (4), 770-779CODEN: CELLB5; ISSN:0092-8674. (Cell Press)T cells can be re-directed to kill cancer cells using chimeric antigen receptors (CARs) or T cell receptors (TCRs). This approach, however, is constrained by the rarity of tumor-specific single antigens. Targeting antigens also found on bystander tissues can cause life-threatening adverse effects. A powerful way to enhance ON-target activity of therapeutic T cells is to engineer them to require combinatorial antigens. Here, the authors engineer a combinatorially activated T cell circuit in which a synthetic Notch receptor for one antigen induces the expression of a CAR for a second antigen. These dual-receptor AND-gate T cells are only armed and activated in the presence of dual antigen tumor cells. These T cells show precise therapeutic discrimination in vivo-sparing single antigen "bystander" tumors while efficiently clearing combinatorial antigen "disease" tumors. This type of precision dual-receptor circuit opens the door to immune recognition of a wider range of tumors.
- 20Esensten, J. H.; Bluestone, J. A.; Lim, W. A. Engineering Therapeutic T Cells: From Synthetic Biology to Clinical Trials. Annu. Rev. Pathol 2017, 12, 305– 330, DOI: 10.1146/annurev-pathol-052016-10030420https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVyis7vN&md5=601808024db94dc019410b9b0026bdeaEngineering Therapeutic T Cells: From Synthetic Biology to Clinical TrialsEsensten, Jonathan H.; Bluestone, Jeffrey A.; Lim, Wendell A.Annual Review of Pathology: Mechanisms of Disease (2017), 12 (), 305-330CODEN: ARPMCU; ISSN:1553-4006. (Annual Reviews)Engineered T cells are currently in clin. trials to treat patients with cancer, solid organ transplants, and autoimmune diseases. However, the field is still in its infancy. The design, and manufg., of T cell therapies is not standardized and is performed mostly in academic settings by competing groups. Reliable methods to define dose and pharmacokinetics of T cell therapies need to be developed. As of mid-2016, there are no US Food and Drug Administration (FDA)-approved T cell therapeutics on the market, and FDA regulations are only slowly adapting to the new technologies. Further development of engineered T cell therapies requires advances in immunol., synthetic biol., manufg. processes, and government regulation. In this review, we outline some of these challenges and discuss the contributions that pathologists can make to this emerging field.
- 21Cho, J. H.; Collins, J. J.; Wong, W. W. Universal Chimeric Antigen Receptors for Multiplexed and Logical Control of T Cell Responses. Cell 2018, 173 (6), 1426– 1438.e11, DOI: 10.1016/j.cell.2018.03.03821https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXosFWhsLs%253D&md5=4e6791c20148e208a9eb9ac031d351eaUniversal Chimeric Antigen Receptors for Multiplexed and Logical Control of T Cell ResponsesCho, Jang Hwan; Collins, James J.; Wong, Wilson W.Cell (Cambridge, MA, United States) (2018), 173 (6), 1426-1438.e11CODEN: CELLB5; ISSN:0092-8674. (Cell Press)T cells expressing chimeric antigen receptors (CARs) are promising cancer therapeutic agents, with the prospect of becoming the ultimate smart cancer therapeutics. To expand the capability of CAR T cells, here, we present a split, universal, and programmable (SUPRA) CAR system that simultaneously encompasses multiple crit. "upgrades," such as the ability to switch targets without re-engineering the T cells, finely tune T cell activation strength, and sense and logically respond to multiple antigens. These features are useful to combat relapse, mitigate over-activation, and enhance specificity. We test our SUPRA system against two different tumor models to demonstrate its broad utility and humanize its components to minimize potential immunogenicity concerns. Furthermore, we extend the orthogonal SUPRA CAR system to regulate different T cell subsets independently, demonstrating a dually inducible CAR system. Together, these SUPRA CARs illustrate that multiple advanced logic and control features can be implemented into a single, integrated system.
- 22Kudo, K.; Imai, C.; Lorenzini, P.; Kamiya, T.; Kono, K.; Davidoff, A.; Chng, W.; Campana, D. T lymphocytes expressing a CD16 signaling receptor exert antibody-dependent cancer cell killing. Cancer Res. 2014, 74 (1), 93– 103, DOI: 10.1158/0008-5472.CAN-13-136522https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXislKhtQ%253D%253D&md5=997922da6b258eb409360d48e8bdcbb9T Lymphocytes Expressing a CD16 Signaling Receptor Exert Antibody-Dependent Cancer Cell KillingKudo, Ko; Imai, Chihaya; Lorenzini, Paolo; Kamiya, Takahiro; Kono, Koji; Davidoff, Andrew M.; Chng, Wee Joo; Campana, DarioCancer Research (2014), 74 (1), 93-103CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)To expand applications for T-cell-based immunotherapy in cancer, we designed a receptor that binds the Fc portion of human Igs and delivers activation signals. The construct included the high-affinity CD16 (FCGR3A) V158 variant, CD8α hinge, and transmembrane domains, along with signaling domains from CD3ζ and 4-1BB (TNFRSF9), forming a chimeric receptor termed CD16V-BB-ζ After retrovirus-mediated expression in human T cells, CD16V-BB-ζ- bound humanized antibodies with higher affinity than a control receptor contg. the more common F158 variant. Engagement of CD16V-VV-ζ provoked T-cell activation, exocytosis of lytic granules, and sustained proliferation, with a mean cell recovery after 4-wk coculture with Daudi lymphoma cells and rituximab of nearly 70-fold relative to input cells. In contrast, unbound antibody alone produced no effect. CD16-BB-ζ T cells specifically killed lymphoma cells and primary chronic lymphocytic leukemia cells in combination with rituximab at a low effectontarget ratio, even when assayed on mesenchymal cells. Trastuzumab triggered CD16V-BB-ζ-mediated killing of HER2 (ERBB2)+ breast and gastric cancer cells; similar results were obtained with an anti-GD2 antibody in neuroblastoma and osteosarcoma cells. Furthermore, coadministration of CD16V-BB-ζ T cells with immunotherapeutic antibodies exerted considerable antitumor activity in vivo. Signaling mediated by 4-1BB-CD3ζ induced higher T-cell activation, proliferation, and cytotoxicity than 3 or FcεRIγ, and the receptor was expressed effectively after mRNA electroporation without viral vectors, facilitating clin. translation. Our results offer preclin. proof of concept for CD16V-BB-ζ as a universal, next-generation chimeric receptor with the potential to augment the efficacy of antibody therapies for cancer.
- 23Minutolo, N. G.; Hollander, E. E.; Powell, D. J., Jr The Emergence of Universal Immune Receptor T Cell Therapy for Cancer. Front. Oncol. 2019, 9, 176, DOI: 10.3389/fonc.2019.0017623https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M%252Fltl2qsA%253D%253D&md5=05413c83ff8ab6c6fddacd1b16d7c2d9The Emergence of Universal Immune Receptor T Cell Therapy for CancerMinutolo Nicholas G; Hollander Erin E; Powell Daniel J Jr; Minutolo Nicholas G; Minutolo Nicholas G; Minutolo Nicholas G; Hollander Erin E; Powell Daniel J Jr; Hollander Erin EFrontiers in oncology (2019), 9 (), 176 ISSN:2234-943X.Chimeric antigen receptor (CAR) T cells have shown great success in the treatment of CD19+ hematological malignancies, leading to their recent approval by the FDA as a new cancer treatment modality. However, their broad use is limited since a CAR targets a single tumor associated antigen (TAA), which is not effective against tumors with heterogeneous TAA expression or emerging antigen loss variants. Further, stably engineered CAR T cells can continually and uncontrollably proliferate and activate in response to antigen, potentially causing fatal on-target off-tumor toxicity, cytokine release syndrome, or neurotoxicity without a method of control or elimination. To address these issues, our lab and others have developed various universal immune receptors (UIRs) that allow for targeting of multiple TAAs by T cells expressing a single receptor. UIRs function through the binding of an extracellular adapter domain which acts as a bridge between intracellular T cell signaling domains and a soluble tumor antigen targeting ligand (TL). The dissociation of TAA targeting and T cell signaling confers many advantages over standard CAR therapy, such as dose control of T cell effector function, the ability to simultaneously or sequentially target multiple TAAs, and control of immunologic synapse geometry. There are currently four unique UIR platform types: ADCC-mediating Fc-binding immune receptors, bispecific protein engaging immune receptors, natural binding partner immune receptors, and anti-tag CARs. These UIRs all allow for potential benefits over standard CARs, but also bring unique engineering challenges that will have to be addressed to achieve maximal efficacy and safety in the clinic. Still, UIRs present an exciting new avenue for adoptive T cell transfer therapies and could lead to their expanded use in areas which current CAR therapies have failed. Here we review the development of each UIR platform and their unique functional benefits, and detail the potential hurdles that may need to be overcome for continued clinical translation.
- 24Ruffo, E.; Butchy, A. A.; Tivon, Y.; So, V.; Kvorjak, M.; Parikh, A.; Adams, E. L.; Miskov-Zivanov, N.; Finn, O. J.; Deiters, A. Post-translational covalent assembly of CAR and synNotch receptors for programmable antigen targeting. Nat. Commun. 2023, 14 (1), 2463, DOI: 10.1038/s41467-023-37863-524https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXpsl2iurY%253D&md5=5f0895c2abd463a1ea1ce95ae02196a5Post-translational covalent assembly of CAR and synNotch receptors for programmable antigen targetingRuffo, Elisa; Butchy, Adam A.; Tivon, Yaniv; So, Victor; Kvorjak, Michael; Parikh, Avani; Adams, Eric L.; Miskov-Zivanov, Natasa; Finn, Olivera J.; Deiters, Alexander; Lohmueller, JasonNature Communications (2023), 14 (1), 2463CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)Chimeric antigen receptors (CARs) and synthetic Notch (synNotch) receptors are engineered cell-surface receptors that sense a target antigen and respond by activating T cell receptor signaling or a customized gene program, resp. Here, to expand the targeting capabilities of these receptors, we develop "universal" receptor systems for which receptor specificity can be directed post-translationally via covalent attachment of a co-administered antibody bearing a benzylguanine (BG) motif. A SNAPtag self-labeling enzyme is genetically fused to the receptor and reacts with BG-conjugated antibodies for covalent assembly, programming antigen recognition. We demonstrate that activation of SNAP-CAR and SNAP-synNotch receptors can be successfully targeted by clin. relevant BG-conjugated antibodies, including anti-tumor activity of SNAP-CAR T cells in vivo in a human tumor xenograft mouse model. Finally, we develop a math. model to better define the parameters affecting universal receptor signaling. SNAP receptors provide a powerful strategy to post-translationally reprogram the targeting specificity of engineered cells.
From NLM Medline
- 25Lohmueller, J. J.; Ham, J. D.; Kvorjak, M.; Finn, O. J. mSA2 affinity-enhanced biotin-binding CAR T cells for universal tumor targeting. Oncoimmunology 2018, 7 (1), e1368604 DOI: 10.1080/2162402X.2017.136860425https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhslaltL%252FJ&md5=011672c41c0ebb99f473cd3328f9dfc3mSA2 affinity-enhanced biotin-binding CAR T cells for universal tumor targetingLohmueller, Jason J.; Ham, James D.; Kvorjak, Michael; Finn, Olivera J.OncoImmunology (2018), 7 (1), e1368604/1-e1368604/6CODEN: ONCOGX; ISSN:2162-402X. (Taylor & Francis, Inc.)Chimeric antigen receptor T cells (CAR-Ts) are promising cancer therapeutics. However, since cancer cells can lose the CAR-targeted antigen and avoid destruction, targeting multiple antigens with multiple CARs has been proposed. We illustrate here a less cumbersome alternative, anti-tag CARs (AT-CARs) that bind to tags on tumor-targeting antibodies. We have created novel AT-CARs, using the affinity-enhanced monomeric streptavidin 2 (mSA2) biotin-binding domain that when expressed on T cells can target cancer cells coated with biotinylated antibodies. Human T cells expressing mSA2 CARs with CD28-CD3ζ and 4-1BB-CD3ζ signaling domains were activated by plate-immobilized biotin and by tumor cells coated with biotinylated antibodies against the tumor-assocd. antigens CD19 and CD20. Furthermore, mSA2 CAR T cells were capable of mediating cancer cell lysis and IFNγ prodn. in an antibody dose-dependent manner. The mSA2 CAR is a universal AT-CAR that can be combined with biotinylated tumor-specific antibodies to potentially target many different tumor types.
- 26Tamada, K.; Geng, D.; Sakoda, Y.; Bansal, N.; Srivastava, R.; Li, Z.; Davila, E. Redirecting gene-modified T cells toward various cancer types using tagged antibodies. Clin. Cancer Res. 2012, 18 (23), 6436– 6445, DOI: 10.1158/1078-0432.CCR-12-144926https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhslKgurjJ&md5=93e3532e765309253195922cc35ea078Redirecting Gene-Modified T Cells toward Various Cancer Types Using Tagged AntibodiesTamada, Koji; Geng, Degui; Sakoda, Yukimi; Bansal, Navneeta; Srivastava, Ratika; Li, Zhaoyang; Davila, EduardoClinical Cancer Research (2012), 18 (23), 6436-6445CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: To develop an adaptable gene-based vector that will confer immune cell specificity to various cancer types. Exptl. Design: Human and mouse T cells were genetically engineered to express a chimeric antigen receptor (CAR) that binds a fluorescein isothiocyanate (FITC) mol., termed anti-FITC CAR T cells. Various antibodies (Ab) currently in clin. use including cetuximab (Ctx), trastuzumab (Her2), and rituximab (Rtx) were conjugated with FITC and tested for their ability to bind tumor cells, activate T cells, and induce antitumor effects in vitro and in vivo. Results: Anti-FITC CAR T cells recognize various cancer types when bound with FITC-labeled Abs resulting in efficient target lysis, T-cell proliferation, and cytokine/chemokine prodn. The treatment of immunocompromised mice with human anti-FITC CAR T cells plus FITC-labeled cetuximab (FITC-Ctx) delayed the growth of colon cancer but unexpectedly led to the outgrowth of EGF receptor (EGFR)-neg. tumor cells. On the other hand, in a human pancreatic cancer cell line with uniform EGFR expression, anti-FITC CAR T cells plus FITC-Ctx eradicated preestablished late-stage tumors. In immunocompetent mice, anti-FITC CAR T cells exhibited potent antitumor activity against syngeneic mouse breast cancer expressing Her2 and B-cell lymphoma expressing CD20 by combining with FITC-Her2 and FITC-Rtx, resp. In addn., the activity of anti-FITC CAR T cells could be attenuated by subsequent injections of nonspecific FITC-IgG. Conclusion: These studies highlight an applicability of anti-tag CAR technol. to treat patients with different types of cancers and a possibility to regulate CAR T-cell functions with competing FITC mols.
- 27Ma, J. S.; Kim, J. Y.; Kazane, S. A.; Choi, S. H.; Yun, H. Y.; Kim, M. S.; Rodgers, D. T.; Pugh, H. M.; Singer, O.; Sun, S. B. Versatile strategy for controlling the specificity and activity of engineered T cells. Proc. Natl. Acad. Sci. U.S.A. 2016, 113 (4), E450– E458, DOI: 10.1073/pnas.152419311327https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xns1antA%253D%253D&md5=8a3ca3ccfac0d66d05ec4be93323a970Versatile strategy for controlling the specificity and activity of engineered T cellsMa, Jennifer S. Y.; Kim, Ji Young; Kazane, Stephanie A.; Choi, Sei-hyun; Yun, Hwa Young; Kim, Min Soo; Rodgers, David T.; Pugh, Holly M.; Singer, Oded; Sun, Sophie B.; Fonslow, Bryan R.; Kochenderfer, James N.; Wright, Timothy M.; Schultz, Peter G.; Young, Travis S.; Kim, Chan Hyuk; Cao, YuProceedings of the National Academy of Sciences of the United States of America (2016), 113 (4), E450-E458CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The adoptive transfer of autologous T cells engineered to express a chimeric antigen receptor (CAR) has emerged as a promising cancer therapy. Despite impressive clin. efficacy, the general application of current CAR-T-cell therapy is limited by serious treatment-related toxicities. One approach to improve the safety of CAR-T cells involves making their activation and proliferation dependent upon adaptor mols. that mediate formation of the immunol. synapse between the target cancer cell and T-cell. Here, the authors describe the design and synthesis of structurally defined semisynthetic adaptors the authors refer to as "switch" mols., in which anti-CD19 and anti-CD22 antibody fragments are site-specifically modified with FITC using genetically encoded noncanonical amino acids. This approach allows the precise control over the geometry and stoichiometry of complex formation between CD19- or CD22-expressing cancer cells and a "universal" anti-FITC-directed CAR-T cell. Optimization of this CAR-switch combination results in potent, dose-dependent in vivo antitumor activity in xenograft models. The advantage of being able to titrate CAR-T-cell in vivo activity was further evidenced by reduced in vivo toxicity and the elimination of persistent B-cell aplasia in immune-competent mice. The ability to control CAR-T cell and cancer cell interactions using intermediate switch mols. may expand the scope of engineered T-cell therapy to solid tumors, as well as indications beyond cancer therapy.
- 28Rodgers, D. T.; Mazagova, M.; Hampton, E. N.; Cao, Y.; Ramadoss, N. S.; Hardy, I. R.; Schulman, A.; Du, J.; Wang, F.; Singer, O. Switch-mediated activation and retargeting of CAR-T cells for B-cell malignancies. Proc. Natl. Acad. Sci. U.S.A. 2016, 113 (4), E459– E468, DOI: 10.1073/pnas.152415511328https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xns1antw%253D%253D&md5=a4d4b98acb1877a0394582a6b5bd374cSwitch-mediated activation and retargeting of CAR-T cells for B-cell malignanciesRodgers, David T.; Mazagova, Magdalena; Hampton, Eric N.; Cao, Yu; Ramadoss, Nitya S.; Hardy, Ian R.; Schulman, Andrew; Du, Juanjuan; Wang, Feng; Singer, Oded; Ma, Jennifer; Nunez, Vanessa; Shen, Jiayin; Woods, Ashley K.; Wright, Timothy M.; Schultz, Peter G.; Kim, Chan Hyuk; Young, Travis S.Proceedings of the National Academy of Sciences of the United States of America (2016), 113 (4), E459-E468CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Chimeric antigen receptor T (CAR-T) cell therapy has produced impressive results in clin. trials for B-cell malignancies. However, safety concerns related to the inability to control CAR-T cells once infused into the patient remain a significant challenge. Here the authors report the engineering of recombinant antibody-based bifunctional switches that consist of a tumor antigen-specific Fab mol. engrafted with a peptide neo-epitope, which is bound exclusively by a peptide-specific switchable CAR-T cell (sCAR-T). The switch redirects the activity of the bio-orthogonal sCAR-T cells through the selective formation of immunol. synapses, in which the sCAR-T cell, switch, and target cell interact in a structurally defined and temporally controlled manner. Optimized switches specific for CD19 controlled the activity, tissue-homing, cytokine release, and phenotype of sCAR-T cells in a dose-titratable manner in a Nalm-6 xenograft rodent model of B-cell leukemia. The sCAR-T-cell dosing regimen could be tuned to provide efficacy comparable to the corresponding conventional CART-19, but with lower cytokine levels, thereby offering a method of mitigating cytokine release syndrome in clin. translation. Furthermore, the authors demonstrate that this methodol. is readily adaptable to targeting CD20 on cancer cells using the same sCAR-T cell, suggesting that this approach may be broadly applicable to heterogeneous and resistant tumor populations, as well as other liq. and solid tumor antigens.
- 29Lamers, C. H.; Sleijfer, S.; van Steenbergen, S.; van Elzakker, P.; van Krimpen, B.; Groot, C.; Vulto, A.; den Bakker, M.; Oosterwijk, E.; Debets, R. Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol. Ther. 2013, 21 (4), 904– 912, DOI: 10.1038/mt.2013.1729https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXislWhtbw%253D&md5=c6a35182968904a9deb6a7d0890071dcTreatment of Metastatic Renal Cell Carcinoma With CAIX CAR-engineered T cells: Clinical Evaluation and Management of On-target ToxicityLamers, Cor H. J.; Sleijfer, Stefan; van Steenbergen, Sabine; van Elzakker, Pascal; van Krimpen, Brigitte; Groot, Corrien; Vulto, Arnold; den Bakker, Michael; Oosterwijk, Egbert; Debets, Reno; Gratama, Jan W.Molecular Therapy (2013), 21 (4), 904-912CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)Autologous T cells genetically modified to express a chimeric antibody receptor (CAR) against carboxy-anhydrase-IX (CAIX) were administered to 12 patients with CAIX-expressing metastatic renal cell carcinoma (RCC). Patients were treated in three cohorts with a max. of 10 infusions of a total of 0.2 to 2.1 × 109 CAR T cells. CTC grade 2-4 liver enzyme disturbances occurred at the lowest CAR T cell doses, necessitating cessation of treatment in four out of eight patients in cohorts 1 and 2. Examn. of liver biopsies revealed CAIX expression on bile duct epithelium with infiltration of T cells, including CAR T cells. Subsequently four patients were pre-treated with CAIX monoclonal antibody (mAb) G250 to prevent CAR-specific toxicity and showed no liver toxicities and indications for enhanced peripheral T cell persistence. No clin. responses were recorded. This report shows that CAIX-targeting CAR T cells exerted antigen-specific effects in vivo and induced liver toxicity at the lowest dose of 0.2 × 109 T cells applied, illustrating the potency of receptor-modified T cells. We provide in-patient proof that the obsd. "on-target" toxicity is antigen-directed and can be prevented by blocking antigenic sites in off-tumor organs and allowing higher T cell doses.
- 30Morgan, R. A.; Yang, J. C.; Kitano, M.; Dudley, M. E.; Laurencot, C. M.; Rosenberg, S. A. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 2010, 18 (4), 843– 851, DOI: 10.1038/mt.2010.2430https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXitlaitb4%253D&md5=fdf1fd771d8ce6bb9c83f46e58d89db6Case Report of a Serious Adverse Event Following the Administration of T Cells Transduced With a Chimeric Antigen Receptor Recognizing ERBB2Morgan, Richard A.; Yang, James C.; Kitano, Mio; Dudley, Mark E.; Laurencot, Carolyn M.; Rosenberg, Steven A.Molecular Therapy (2010), 18 (4), 843-851CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)In an attempt to treat cancer patients with ERBB2 overexpressing tumors, we developed a chimeric antigen receptor (CAR) based on the widely used humanized monoclonal antibody (mAb) Trastuzumab (Herceptin). An optimized CAR vector contg. CD28, 4-1BB, and CD3ζ signaling moieties was assembled in a γ-retroviral vector and used to transduce autologous peripheral blood lymphocytes (PBLs) from a patient with colon cancer metastatic to the lungs and liver, refractory to multiple std. treatments. The gene transfer efficiency into autologous T cells was 79% CAR+ in CD3+ cells and these cells demonstrated high-specific reactivity in in vitro coculture assays. Following completion of nonmyeloablative conditioning, the patient received 1010 cells i.v. Within 15 min after cell infusion the patient experienced respiratory distress, and displayed a dramatic pulmonary infiltrate on chest X-ray. She was intubated and despite intensive medical intervention the patient died 5 days after treatment. Serum samples after cell infusion showed marked increases in interferon-γ (IFN-γ), granulocyte macrophage-colony stimulating factor (GM-CSF), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-10, consistent with a cytokine storm. We speculate that the large no. of administered cells localized to the lung immediately following infusion and were triggered to release cytokine by the recognition of low levels of ERBB2 on lung epithelial cells.
- 31Gardner, L.; Deiters, A. Light-controlled synthetic gene circuits. Curr. Opin. Chem. Biol. 2012, 16 (3–4), 292– 299, DOI: 10.1016/j.cbpa.2012.04.01031https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XnslSitro%253D&md5=1e2bc100aebe766aed22d2937bba8f47Light-controlled synthetic gene circuitsGardner, Laura; Deiters, AlexanderCurrent Opinion in Chemical Biology (2012), 16 (3-4), 292-299CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. Highly complex synthetic gene circuits have been engineered in living organisms to develop systems with new biol. properties. A precise trigger to activate or deactivate these complex systems is desired in order to tightly control different parts of a synthetic or natural network. Light represents an excellent tool to achieve this goal as it can be regulated in timing, location, intensity, and wavelength, which allows for precise spatiotemporal control over genetic circuits. Recently, light has been used as a trigger to control the biol. function of small mols., oligonucleotides, and proteins involved as parts in gene circuits. Light activation has enabled the construction of unique systems in living organisms such as band-pass filters and edge-detectors in bacterial cells. Addnl., light also allows for the regulation of intermediate steps of complex dynamic pathways in mammalian cells such as those involved in kinase networks. Herein we describe recent advancements in the area of light-controlled synthetic networks.
From NLM Medline
- 32Ankenbruck, N.; Courtney, T.; Naro, Y.; Deiters, A. Optochemical Control of Biological Processes in Cells and Animals. Angew. Chem., Int. Ed. Engl. 2018, 57 (11), 2768– 2798, DOI: 10.1002/anie.20170017132https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1crovFGnsg%253D%253D&md5=8f1ab7383a0629b0ed87b58d47ad0727Optochemical Control of Biological Processes in Cells and AnimalsAnkenbruck Nicholas; Courtney Taylor; Naro Yuta; Deiters AlexanderAngewandte Chemie (International ed. in English) (2018), 57 (11), 2768-2798 ISSN:.Biological processes are naturally regulated with high spatial and temporal control, as is perhaps most evident in metazoan embryogenesis. Chemical tools have been extensively utilized in cell and developmental biology to investigate cellular processes, and conditional control methods have expanded applications of these technologies toward resolving complex biological questions. Light represents an excellent external trigger since it can be controlled with very high spatial and temporal precision. To this end, several optically regulated tools have been developed and applied to living systems. In this review we discuss recent developments of optochemical tools, including small molecules, peptides, proteins, and nucleic acids that can be irreversibly or reversibly controlled through light irradiation, with a focus on applications in cells and animals.
From NLM Medline
- 33Davies, S.; Stenton, B. J.; Bernardes, G. J. L. Bioorthogonal Decaging Reactions for Targeted Drug Activation. Chimia 2018, 72 (11), 771– 776, DOI: 10.2533/chimia.2018.77133https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvVaqsLs%253D&md5=21cc53fef93345d64f1c9351b9c83debBioorthogonal decaging reactions for targeted drug activationDavies, Sarah; Stenton, Benjamin J.; Bernardes, Goncalo J. L.Chimia (2018), 72 (11), 771-776CODEN: CHIMAD ISSN:. (Swiss Chemical Society)Bioorthogonal decaging reactions are highly selective transformations which involve the cleavage of a protecting group from a mol. of interest. Decaging reactions can be classified into subgroups depending on the nature of the trigger; they can be photo-, metal- or small mol.-triggered. Due to their highly selective and biocompatible nature, they can be carried out in living systems as they do not interfere with any endogenous processes. This gain-of-function allows controlled activation of proteins and release of fluorophores and drugs in vivo. Although there are many examples of fluorophore/protein release, this review focuses on the application of bioorthogonal decaging reactions for targeted drug activation. One strategy for targeted drug delivery is tissue-selective activation of prodrugs and antibody-drug conjugates (ADCs). Bioorthogonal decaging provides a highly selective, controllable method for activating prodrugs and ADCs, reducing toxicity due to the off-target drug release that occurs in endogenous activation strategies. Here we focus on the development of bifunctional linkers that enable studies of bioorthogonal chem. for activation of ADCs.
From NLM Medline
- 34van der Gracht, A. M. F.; de Geus, M. A. R.; Camps, M. G. M.; Ruckwardt, T. J.; Sarris, A. J. C.; Bremmers, J.; Maurits, E.; Pawlak, J. B.; Posthoorn, M. M.; Bonger, K. M. Chemical Control over T-Cell Activation in Vivo Using Deprotection of trans-Cyclooctene-Modified Epitopes. ACS Chem. Biol. 2018, 13 (6), 1569– 1576, DOI: 10.1021/acschembio.8b0015534https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MfgsVKrtA%253D%253D&md5=e2ae7a8dc3a10d411afcd5873a596eadChemical Control over T-Cell Activation in Vivo Using Deprotection of trans-Cyclooctene-Modified Epitopesvan der Gracht Anouk M F; de Geus Mark A R; Sarris Alexi J C; Bremmers Jessica; Maurits Elmer; Pawlak Joanna B; Posthoorn Michelle M; Filippov Dmitri V; Overkleeft Herman S; van Kasteren Sander I; Camps Marcel G M; Ossendorp Ferry; Ruckwardt Tracy J; Bonger Kimberly M; Robillard Marc SACS chemical biology (2018), 13 (6), 1569-1576 ISSN:.Activation of a cytotoxic T-cell is a complex multistep process, and tools to study the molecular events and their dynamics that result in T-cell activation in situ and in vivo are scarce. Here, we report the design and use of conditional epitopes for time-controlled T-cell activation in vivo. We show that trans-cyclooctene-protected SIINFEKL (with the lysine amine masked) is unable to elicit the T-cell response characteristic for the free SIINFEKL epitope. Epitope uncaging by means of an inverse-electron demand Diels-Alder (IEDDA) event restored T-cell activation and provided temporal control of T-cell proliferation in vivo.
From NLM Medline
- 35Li, J.; Chen, P. R. Development and application of bond cleavage reactions in bioorthogonal chemistry. Nat. Chem. Biol. 2016, 12 (3), 129– 137, DOI: 10.1038/nchembio.202435https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XivVCis74%253D&md5=6b563f0c618e43f97bf4cea26c060451Development and application of bond cleavage reactions in bioorthogonal chemistryLi, Jie; Chen, Peng R.Nature Chemical Biology (2016), 12 (3), 129-137CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)A review. Bioorthogonal chem. reactions are a thriving area of chem. research in recent years as an unprecedented technique to dissect native biol. processes through chem.-enabled strategies. However, current concepts of bioorthogonal chem. have largely centered on bond formation reactions between two mutually reactive bioorthogonal handles. Recently, in a reverse strategy, a collection of bond cleavage reactions has emerged with excellent biocompatibility. These reactions have expanded the bioorthogonal chem. repertoire, enabling an array of exciting new biol. applications that range from the chem. controlled spatial and temporal activation of intracellular proteins and small-mol. drugs to the direct manipulation of intact cells under physiol. conditions. Here the authors highlight the development and applications of these bioorthogonal cleavage reactions. Furthermore, the authors lay out challenges and propose future directions along this appealing avenue of research.
From NLM Medline
- 36Scinto, S. L.; Bilodeau, D. A.; Hincapie, R.; Lee, W.; Nguyen, S. S.; Xu, M.; Am Ende, C. W.; Finn, M. G.; Lang, K.; Lin, Q. Bioorthogonal chemistry. Nat. Rev. Methods Primers 2021, 1, 30, DOI: 10.1038/s43586-021-00028-z36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XjsVGgu7k%253D&md5=64eb15e12374df94c7b27bc4c9ef2accBioorthogonal chemistryScinto, Samuel L.; Bilodeau, Didier A.; Hincapie, Robert; Lee, Wankyu; Nguyen, Sean S.; Xu, Minghao; am Ende, Christopher W.; Finn, M. G.; Lang, Kathrin; Lin, Qing; Pezacki, John Paul; Prescher, Jennifer A.; Robillard, Marc S.; Fox, Joseph M.Nature Reviews Methods Primers (2021), 1 (1), 30CODEN: NRMPAT; ISSN:2662-8449. (Nature Portfolio)A review. Bioorthogonal chem. represents a class of high-yielding chem. reactions that proceed rapidly and selectively in biol. environments without side reactions towards endogenous functional groups. Rooted in the principles of phys. org. chem., bioorthogonal reactions are intrinsically selective transformations not commonly found in biol. Key reactions include native chem. ligation and the Staudinger ligation, copper-catalyzed azide-alkyne cycloaddn., strain-promoted [3 + 2] reactions, tetrazine ligation, metal-catalyzed coupling reactions, oxime and hydrazone ligations as well as photoinducible bioorthogonal reactions. Bioorthogonal chem. has significant overlap with the broader field of click chem. - high-yielding reactions that are wide in scope and simple to perform, as recently exemplified by sulfuryl fluoride exchange chem. The underlying mechanisms of these transformations and their optimal conditions are described in this Primer, followed by discussion of how bioorthogonal chem. has become essential to the fields of biomedical imaging, medicinal chem., protein synthesis, polymer science, materials science and surface science. The applications of bioorthogonal chem. are diverse and include genetic code expansion and metabolic engineering, drug target identification, antibody-drug conjugation and drug delivery. This Primer describes stds. for reproducibility and data deposition, outlines how current limitations are driving new research directions and discusses new opportunities for applying bioorthogonal chem. to emerging problems in biol. and biomedicine.
- 37Tu, J.; Xu, M.; Franzini, R. M. Dissociative Bioorthogonal Reactions. ChemBioChem 2019, 20 (13), 1615– 1627, DOI: 10.1002/cbic.20180081037https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXotFamsbY%253D&md5=e9277eac67c6092b293c936477e47467Dissociative Bioorthogonal ReactionsTu, Julian; Xu, Minghao; Franzini, Raphael M.ChemBioChem (2019), 20 (13), 1615-1627CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)Bioorthogonal reactions that proceed readily under physiol. conditions without interference from biomols. have found widespread application in the life sciences. Complementary to the bioorthogonal reactions that ligate two mols., reactions that release a mol. or cleave a linker are increasingly attracting interest. Such dissociative bioorthogonal reactions have a broad spectrum of uses, for example, in controlling bio-macromol. activity, in drug delivery, and in diagnostic assays. This review article summarizes the developed bioorthogonal reactions linked to a release step, outlines representative areas of the applications of such reactions, and discusses aspects that require further improvement.
From NLM Medline
- 38Messmann, H.; Endlicher, E.; Freunek, G.; Rummele, P.; Scholmerich, J.; Knuchel, R. Fluorescence endoscopy for the detection of low and high grade dysplasia in ulcerative colitis using systemic or local 5-aminolaevulinic acid sensitisation. Gut 2003, 52 (7), 1003– 1007, DOI: 10.1136/gut.52.7.100338https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3s3nsFantQ%253D%253D&md5=19bd79f280aeb54b5a1a4dac1f95490dFluorescence endoscopy for the detection of low and high grade dysplasia in ulcerative colitis using systemic or local 5-aminolaevulinic acid sensitisationMessmann H; Endlicher E; Freunek G; Rummele P; Scholmerich J; Knuchel RGut (2003), 52 (7), 1003-7 ISSN:0017-5749.BACKGROUND AND AIMS: Longstanding ulcerative colitis (UC), especially in the presence of epithelial dysplasia, is associated with an increased risk of developing cancer. As dysplasia is not visible during routine endoscopy, at least 40-50 random biopsies in four quadrants every 10 cm are recommended. Fluorescence endoscopy after sensitisation with 5-aminolaevulinic acid (5-ALA) was assessed for the detection of dysplasia in ulcerative colitis by taking optical guided biopsies. 5-ALA is converted intracellularly into the sensitiser protoporphyrin IX which accumulates selectively in neoplastic tissue allowing the detection of dysplasia by typical red fluorescence after illumination with blue light. METHODS: In 37 patients with UC, 54 examinations were performed with fluorescence endoscopy after oral (20 mg/kg) or local (either with an enema or by spraying the mucosa with a catheter) sensitisation with 5-ALA. A total of 481 biopsies of red fluorescent (n=218) and non-fluorescent (n=263) areas of the colonic mucosa were taken. RESULTS: Forty two biopsies in 12 patients revealed either low grade (n=40) or high grade (n=2) dysplasia. Sensitivity of fluorescence for dysplastic lesions was excellent and ranged from 87% (95% confidence interval (CI) 0.73-1.00) to 100% (95% CI 1.00-1.00) after local sensitisation, in contrast with only 43% (95% CI 0.17-0.69) after systemic administration. Specificity did not differ for both forms of local sensitisation (enema 51% (95% CI 0.44-0.57) and spray catheter 62% (95% CI 0.51-0.73)); after systemic sensitisation specificity was 73% (95% CI 0.69-0.83). Negative predictive values of non-fluorescent mucosa for exclusion of dysplasia were very high; 89% after systemic sensitisation and 98-100% after local sensitisation. Positive predictive values were 13% and 14% after local sensitisation with enema and spray catheter, and 21% after oral sensitisation with 20 mg/kg ALA. The overall number of biopsies per examination was less than five from fluorescent positive areas. CONCLUSION: Fluorescence endoscopy after 5-ALA sensitisation is a possible tool to visualise dysplastic lesions in ulcerative colitis using 5-ALA sensitisation. Local sensitisation is a promising alternative approach compared with systemic administration of 5-ALA. A randomised controlled study is now indicated to compare the efficacy of endoscopic fluorescence detection with the standard technique of four quadrant random biopsies.
- 39Georgianna, W. E.; Lusic, H.; McIver, A. L.; Deiters, A. Photocleavable polyethylene glycol for the light-regulation of protein function. Bioconjugate Chem. 2010, 21 (8), 1404– 1407, DOI: 10.1021/bc100084n39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptFGgur4%253D&md5=117f49d83b85ea19b2f735335ac18184Photocleavable Polyethylene Glycol for the Light-Regulation of Protein FunctionGeorgianna, Wesleigh E.; Lusic, Hrvoje; McIver, Andrew L.; Deiters, AlexanderBioconjugate Chemistry (2010), 21 (8), 1404-1407CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)PEGylation is commonly employed to enhance the pharmacokinetic properties of proteins, but it can interfere with natural protein function. Protein activity can thus be abrogated through PEGylation, and a controllable means to remove the polyethylene glycol (PEG) group from the protein is desirable. As such, light affords a unique control over biomols. through the application of photosensitive groups. Herein, the authors report the synthesis of a photocleavable PEG reagent (PhotoPEG) and its application to the light-regulation of enzyme activity.
From NLM Medline
- 40Govan, J. M.; McIver, A. L.; Deiters, A. Stabilization and photochemical regulation of antisense agents through PEGylation. Bioconjugate Chem. 2011, 22 (10), 2136– 2142, DOI: 10.1021/bc200411n40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1ChtLfO&md5=b0a42ab4adfaee28ac04e19c4ebf2679Stabilization and Photochemical Regulation of Antisense Agents through PEGylationGovan, Jeane M.; McIver, Andrew L.; Deiters, AlexanderBioconjugate Chemistry (2011), 22 (10), 2136-2142CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Oligonucleotides are effective tools for the regulation of gene expression in cell culture and model organisms, most importantly through antisense mechanisms. Due to the inherent instability of DNA antisense agents, various modifications have been introduced to increase the efficacy of oligonucleotides, including phosphorothioate DNA, locked nucleic acids, peptide nucleic acids, and others. Here, we present antisense agent stabilization through conjugation of a poly(ethylene glycol) (PEG) group to a DNA oligonucleotide. By employing a photocleavable linker between the PEG group and the antisense agent, we were able to achieve light-induced deactivation of antisense activity. The bioconjugated PEG group provides stability to the DNA antisense agent without affecting its native function of silencing gene expression via RNase H-catalyzed mRNA degrdn. Once irradiated with UV light of 365 nm, the PEG group is cleaved from the antisense agent leaving the DNA unprotected and open for degrdn. by endogenous nucleases, thereby restoring gene expression. By using a photocleavable PEG group (PhotoPEG), antisense activity can be regulated with high spatial and temporal resoln., paving the way for precise regulation of gene expression in biol. systems.
From NLM Medline
- 41Govan, J. M.; Uprety, R.; Thomas, M.; Lusic, H.; Lively, M. O.; Deiters, A. Cellular delivery and photochemical activation of antisense agents through a nucleobase caging strategy. ACS Chem. Biol. 2013, 8 (10), 2272– 2282, DOI: 10.1021/cb400293e41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1Srtr7P&md5=d7442889bc061661f09617e5656092cbCellular Delivery and Photochemical Activation of Antisense Agents through a Nucleobase Caging StrategyGovan, Jeane M.; Uprety, Rajendra; Thomas, Meryl; Lusic, Hrvoje; Lively, Mark O.; Deiters, AlexanderACS Chemical Biology (2013), 8 (10), 2272-2282CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Antisense oligonucleotides are powerful tools to regulate gene expression in cells and model organisms. However, a transfection or microinjection is typically needed for efficient delivery of the antisense agent. We report the conjugation of multiple HIV TAT peptides to a hairpin-protected antisense agent through a light-cleavable nucleobase caging group. This conjugation allows for the facile delivery of the antisense agent without a transfection reagent, and photochem. activation offers precise control over gene expression. The developed approach is highly modular, as demonstrated by the conjugation of folic acid to the caged antisense agent. This enabled targeted cell delivery through cell-surface folate receptors followed by photochem. triggering of antisense activity. Importantly, the presented strategy delivers native oligonucleotides after light-activation, devoid of any delivery functionalities or modifications that could otherwise impair their antisense activity.
From NLM Medline
- 42Yamazoe, S.; Liu, Q.; McQuade, L. E.; Deiters, A.; Chen, J. K. Sequential gene silencing using wavelength-selective caged morpholino oligonucleotides. Angew. Chem., Int. Ed. Engl. 2014, 53 (38), 10114– 10118, DOI: 10.1002/anie.20140535542https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2M%252FisVyitw%253D%253D&md5=3b0e34a24cb4f5fd53ddff365df002b5Sequential gene silencing using wavelength-selective caged morpholino oligonucleotidesYamazoe Sayumi; Liu Qingyang; McQuade Lindsey E; Deiters Alexander; Chen James KAngewandte Chemie (International ed. in English) (2014), 53 (38), 10114-8 ISSN:.Spectrally differentiated caged morpholino oligonucleotides (cMOs) and wavelength-selective illumination have been used to sequentially inactivate organismal gene function. The efficacy of these reverse-genetic chemical probes has been demonstrated in zebrafish embryos, and these reagents have been employed to examine the mechanisms of mesoderm patterning.
From NLM Medline
- 43Brown, W.; Wesalo, J.; Tsang, M.; Deiters, A. Engineering Small Molecule Switches of Protein Function in Zebrafish Embryos. J. Am. Chem. Soc. 2023, 145 (4), 2395– 2403, DOI: 10.1021/jacs.2c1136643https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXhsVeit70%253D&md5=5eca14fb80e49c840a9f63e3da5217a9Engineering Small Molecule Switches of Protein Function in Zebrafish EmbryosBrown, Wes; Wesalo, Joshua; Tsang, Michael; Deiters, AlexanderJournal of the American Chemical Society (2023), 145 (4), 2395-2403CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Precise temporally regulated protein function directs the highly complex processes that make up embryo development. The zebrafish embryo is an excellent model organism to study development, and conditional control over enzymic activity is desirable to target chem. intervention to specific developmental events and to investigate biol. mechanisms. Surprisingly, however, few generally applicable small mol. switches of protein function exist in zebrafish. Genetic code expansion allows for site-specific incorporation of an unnatural amino acids into proteins that contain caging groups that are removed through addn. of small mol. triggers such as phosphines or tetrazines. This broadly applicable control of protein function was applied to activate several enzymes, including a GTPase and a protease, with temporal precision in zebrafish embryos. Simple addn. of the small mol. trigger to the media produces robust and tunable protein activation, which was used to gain insight into the development of a congenital heart defect from a RASopathy mutant of NRAS and to control DNA and protein cleavage events catalyzed by a viral recombinase and a viral protease, resp.
From NLM Medline
- 44Darrah, K.; Wesalo, J.; Lukasak, B.; Tsang, M.; Chen, J. K.; Deiters, A. Small Molecule Control of Morpholino Antisense Oligonucleotide Function through Staudinger Reduction. J. Am. Chem. Soc. 2021, 143 (44), 18665– 18671, DOI: 10.1021/jacs.1c0872344https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitlajur%252FF&md5=2d697c181ecf19a44cc5681fb079b47bSmall Molecule Control of Morpholino Antisense Oligonucleotide Function through Staudinger ReductionDarrah, Kristie; Wesalo, Joshua; Lukasak, Bradley; Tsang, Michael; Chen, James K.; Deiters, AlexanderJournal of the American Chemical Society (2021), 143 (44), 18665-18671CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Conditionally activated, caged morpholino antisense agents (cMOs) are tools that enable the temporal and spatial investigation of gene expression, regulation, and function during embryonic development. Cyclic MOs are conformationally gated oligonucleotide analogs that do not block gene expression until they are linearized through the application of an external trigger, such as light or enzyme activity. Here, we describe the first examples of small mol.-responsive cMOs, which undergo rapid and efficient decaging via a Staudinger redn. This is enabled by a highly flexible linker design that offers opportunities for the installation of chem. activated, self-immolative motifs. We synthesized cyclic cMOs against two distinct, developmentally relevant genes and demonstrated phosphine-triggered knockdown of gene expression in zebrafish embryos. This represents the first report of a small mol.-triggered antisense agent for gene knockdown, adding another bioorthogonal entry to the growing arsenal of gene knockdown tools.
From NLM Medline
- 45Lukasak, B.; Morihiro, K.; Deiters, A. Aryl Azides as Phosphine-Activated Switches for Small Molecule Function. Sci. Rep. 2019, 9 (1), 1470, DOI: 10.1038/s41598-018-37023-645https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cjpvV2mtg%253D%253D&md5=4b9da691d5a6aa1655202d35493630f4Aryl Azides as Phosphine-Activated Switches for Small Molecule FunctionLukasak Bradley; Morihiro Kunihiko; Deiters AlexanderScientific reports (2019), 9 (1), 1470 ISSN:.Engineered small molecule triggers are important tools for the control and investigation of biological processes, in particular protein function. Staudinger reductions of aryl azides to amines through the use of phosphines can trigger an elimination reaction, and thereby activation of a functional molecule, if an appropriately positioned leaving group is present. We conducted detailed investigations of the effect of aryl azide and phosphine structure on both the mechanism and kinetics of these reaction-induced eliminations and identified phosphine/azide pairs that enable complete activation within minutes under physiologically relevant conditions.
From NLM Medline
- 46Wesalo, J. S.; Luo, J.; Morihiro, K.; Liu, J.; Deiters, A. Phosphine-Activated Lysine Analogues for Fast Chemical Control of Protein Subcellular Localization and Protein SUMOylation. ChemBioChem 2020, 21 (1–2), 141– 148, DOI: 10.1002/cbic.20190046446https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitVCkt73L&md5=c43a9a16ad9d11ecacbeb5225570547aPhosphine-Activated Lysine Analogues for Fast Chemical Control of Protein Subcellular Localization and Protein SUMOylationWesalo, Joshua S.; Luo, Ji; Morihiro, Kunihiko; Liu, Jihe; Deiters, AlexanderChemBioChem (2020), 21 (1-2), 141-148CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)The Staudinger redn. and its variants have exceptional compatibility with live cells but can be limited by slow kinetics. Herein we report new small-mol. triggers that turn on proteins through a Staudinger redn./self-immolation cascade with substantially improved kinetics and yields. We achieved this through site-specific incorporation of a new set of azidobenzyloxycarbonyl lysine derivs. in mammalian cells. This approach allowed us to activate proteins by adding a nontoxic, bioorthogonal phosphine trigger. We applied this methodol. to control a post-translational modification (SUMOylation) in live cells, using native modification machinery. This work significantly improves the rate, yield, and tunability of the Staudinger redn.-based activation, paving the way for its application in other proteins and organisms.
From NLM Medline
- 47Lim, K. H.; Huang, H.; Pralle, A.; Park, S. Stable, high-affinity streptavidin monomer for protein labeling and monovalent biotin detection. Biotechnol. Bioeng. 2013, 110 (1), 57– 67, DOI: 10.1002/bit.2460547https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFKlsbjK&md5=62b50d6b861c5cf02ffca311e39acb43Stable, high-affinity streptavidin monomer for protein labeling and monovalent biotin detectionLim, Kok Hong; Huang, Heng; Pralle, Arnd; Park, SheldonBiotechnology and Bioengineering (2013), 110 (1), 57-67CODEN: BIBIAU; ISSN:0006-3592. (John Wiley & Sons, Inc.)The coupling between the quaternary structure, stability and function of streptavidin makes it difficult to engineer a stable, high affinity monomer for biotechnol. applications. For example, the binding pocket of streptavidin tetramer is comprised of residues from multiple subunits, which cannot be replicated in a single domain protein. However, rhizavidin from Rhizobium etli was recently shown to bind biotin with high affinity as a dimer without the hydrophobic tryptophan lid donated by an adjacent subunit. In particular, the binding site of rhizavidin uses residues from a single subunit to interact with bound biotin. The authors therefore postulated that replacing the binding site residues of streptavidin monomer with corresponding rhizavidin residues would lead to the design of a high affinity monomer useful for biotechnol. applications. Here, the authors report the construction and characterization of a structural monomer, mSA, which combines the streptavidin and rhizavidin sequences to achieve optimized biophys. properties. First, the biotin affinity of mSA (Kd = 2.8 nM) is the highest among nontetrameric streptavidin, allowing sensitive monovalent detection of biotinylated ligands. The monomer also has significantly higher stability (Tm = 59.8°) and soly. than all other previously engineered monomers to ensure the mol. remains folded and functional during its application. Using fluorescence correlation spectroscopy, mSA binds biotinylated targets as a monomer. Also the mol. can be used as a genetic tag to introduce biotin binding capability to a heterologous protein. For example, recombinantly fusing the monomer to a cell surface receptor allows direct labeling and imaging of transfected cells using biotinylated fluorophores. A stable and functional streptavidin monomer, such as mSA, should be a useful reagent for designing novel detection systems based on monovalent biotin interaction.
- 48Begovic, M.; Herberman, R. B.; Gorelik, E. Ultraviolet light-induced increase in tumor cell susceptibility to TNF-dependent and TNF-independent natural cell-mediated cytotoxicity. Cell. Immunol. 1991, 138 (2), 349– 359, DOI: 10.1016/0008-8749(91)90159-948https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38XjslWjsA%253D%253D&md5=d2030dbd85b108b7f1081be393a6c4baUltraviolet light-induced increase in tumor cell susceptibility to TNF-dependent and TNF-independent natural cell-mediated cytotoxicityBegovic, Mirsada; Herberman, Ronald B.; Gorelik, ElieserCellular Immunology (1991), 138 (2), 349-59CODEN: CLIMB8; ISSN:0008-8749.Anal. of the effector cells involved in lysis of MCA102 tumor cells was performed by comparing the cytotoxicity of normal spleen cells which mediated both NK and natural cell (NC) cell activity with (a) normal spleen cells in which NC activity was neutralized by anti-TNF Abs (NK+,NC-), (b) NK-depleted or NK-deficient spleen cells (NK-,NC+), and (c) NK-deficient or -depleted spleen cells with NC activity neutralized by anti-TNF Abs (NK-,NC-). Lysis of the original MCA102 tumor cells was relatively low and was mediated by NC cells. UV irradn. increased MCA102 tumor cell sensitivity to lysis by both NK and NC cells. Anal. of the mechanisms involved in UV-induced NK sensitivity revealed that UV irradn. increased tumor cell susceptibility to lytic NK-derived granules. NC sensitivity of MCA102UV tumor cells was assocd. with their increase in sensitivity to TNF and selection of MCA102UV cells for resistance to rTNF resulted in a decrease in their susceptibility to NC cells. To det. how fast UV-induced sensitivity to NCMC and rTNF can be established, 51Cr-labeled MCA102 cells were irradiated in vitro with 38-304 J/m2 of UVC light and their sensitivity to lysis by spleen cells and rTNF was tested immediately in an 18-h cytotoxicity assay. UV treatment with the same doses was repeated 12 days later. The data obtained showed that tumor cell sensitivity to NCMC and TNF appeared shortly after UV irradn., was stable, and was further substantially augmented by the 2nd round of UV treatment. Thus, in vitro UV irradn. of tumor cells could be an effective modulator of tumor cell sensitivity to TNF-dependent and TNF-independent cell-mediated cytotoxicity.
From NLM Medline
- 49Begovic, M.; Herberman, R. B.; Gorelik, E. Effect of UV light on tumor cell sensitivity to NK and NC cell-mediated lysis. Nat. Immun 1993, 12 (4–5), 250– 26649https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXhvVKhtbs%253D&md5=b781014a1179ace46515fd3b502198c3Effect of UV light on tumor cell sensitivity to NK and NC cell-mediated lysisBegovic, Mirsada; Herberman, Ronald B.; Gorelik, ElieserNatural Immunity (1993), 12 (4-5), 250-66CODEN: NAIMEL; ISSN:1018-8916.The effect of UV light irradn. on the immunobiol. properties of murine tumor cells was studied. In vitro irradn. of MCA 102 and MCA 105 fibrosarcomas with a short-wavelength UVC light rendered them highly immunogenic and sensitive to natural cell-mediated cytotoxicity (NCMC). Anal. of the effector cells involved in NCMC revealed that UV irradn. stably increased tumor cell sensitivity to both NK and NC cell lysis. Studies of the mechanisms responsible for increased sensitivity to NK cells indicate that UV treatment did not affect tumor-cell recognition by NK cells but increased their susceptibility to NK-derived lytic granules. Augmentation of UV-treated tumor cell sensitivity to NC cell-mediated lysis was found to be due to their increase in sensitivity to the effector-cell-released TNF. In parallel, UV-treated cells showed high sensitivity to human recombinant TNF whereas untreated parental cells were resistant to rTNF. UV irradn. did not affect rTNF binding, internalization and degrdn. but increased tumor cell vulnerability to TNF-induced DNA fragmentation. Thus, UV light appears as a potent modulator of tumor cell sensitivity to T cell-and natural cell-mediated immunity.
From NLM Medline.
- 50Gorelik, E.; Begovic, M.; Duty, L.; Herberman, R. B. Effect of ultraviolet irradiation on MCA102 tumor cell immunogenicity and sensitivity to tumor necrosis factor. Cancer Res. 1991, 51 (5), 1521– 152850https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXhtl2ju7c%253D&md5=b3d96f18264900670e3e06d0f8710c03Effect of ultraviolet irradiation on MCA102 tumor cell immunogenicity and sensitivity to tumor necrosis factorGorelik, Elieser; Begovic, Mirsada; Duty, Lisa; Herberman, Ronald B.Cancer Research (1991), 51 (5), 1521-8CODEN: CNREA8; ISSN:0008-5472.The ability of UV irradn. to induce immunogenicity of the nonimmunogenic major histocompatibility complex-neg. MCA102 fibrosarcoma was studied. In parallel, the effect of short wavelength UVC light on the sensitivity of tumor cells to natural cell-mediated cytotoxicity and tumor necrosis factor (TNF) was also investigated. MCA102 fibrosarcoma cells were irradiated in vitro twice with UVC light (610 and 457 J/m2). UV treatment changed tumor cell morphol. and increased their in vitro rate of proliferation. However, after inoculation of 1 × 105 to 2 × 106 MCA102UV cells into C57BL/6 mice, growth of these cells was completely prevented. Lyt2.2 and not L3T4 lymphocytes were responsible for the rejection of these tumor cells. After a single dose of UV treatment, tumor growth in C57BL/6 mice was inhibited, particularly with lines irradiated at the highest doses (610 or 457 J/m2). After a 2nd round of irradn., tumor cells became more immunogenic, and the level of tumor growth inhibition increased with higher doses of UV irradn. Thus, cells irradiated twice with 610 and 457 J/m2 became rejectable in all immunocompetent C57BL/6 mice. The increase in tumor cell immunogenicity induced by UV light was not assocd. with the appearance of Class I H-2 antigens. In parallel with the induction of tumor cell immunogenicity, UV irradn. made tumor cells more sensitive to natural cell-mediated cytotoxicity and to lysis by TNF. An increase in sensitivity to natural cell-mediated cytotoxicity and TNF was obsd. after single or double doses (152-610 J/m2) of UV irradn. Thus, UV irradn. appears to be an effective modality for altering tumor cell immunobiol. properties, including increased tumor cell sensitivity to T-cell and/or natural cell-mediated immunity.
From NLM Medline.
- 51Jutz, S.; Leitner, J.; Schmetterer, K.; Doel-Perez, I.; Majdic, O.; Grabmeier-Pfistershammer, K.; Paster, W.; Huppa, J. B.; Steinberger, P. Assessment of costimulation and coinhibition in a triple parameter T cell reporter line: Simultaneous measurement of NF-κB, NFAT and AP-1. J. Immunol. Methods 2016, 430, 10– 20, DOI: 10.1016/j.jim.2016.01.00751https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVKhsb4%253D&md5=3c84198664801ba14aecfb27ba91164cAssessment of costimulation and coinhibition in a triple parameter T cell reporter line: Simultaneous measurement of NF-κB, NFAT and AP-1Jutz, Sabrina; Leitner, Judith; Schmetterer, Klaus; Doel-Perez, Iago; Majdic, Otto; Grabmeier-Pfistershammer, Katharina; Paster, Wolfgang; Huppa, Johannes B.; Steinberger, PeterJournal of Immunological Methods (2016), 430 (), 10-20CODEN: JIMMBG; ISSN:0022-1759. (Elsevier B.V.)Engagement of the T cell receptor complex reprograms T cells for proliferation, cytokine prodn. and differentiation towards effector cells. This process depends on activating costimulatory signals and is counteracted by coinhibitory mols. Three transcription factors, namely NF-κB, NFAT and AP-1, have a major role in inducing the transcriptional program that is required for T cell activation and differentiation. Here we describe the generation of a triple parameter reporter based on the human Jurkat T cell line, where response elements for NF-κB, NFAT and AP-1 drive the expression of the fluorescent proteins CFP, eGFP and mCherry, resp. The emission spectra of these proteins allow simultaneous assessment of NF-κB, NFAT and AP-1 activity in response to stimulation. Ligation of the TCR complex induced moderate reporter activity, which was strongly enhanced upon coengagement of the costimulatory receptors CD2 or CD28. Moreover, we have generated and tested triple parameter reporter cells that harbor costimulatory and inhibitory receptors not endogenously expressed in the Jurkat cells. In these expts. we could show that engagement of the costimulatory mol. 4-1BB enhances NF-κB and AP-1 activity, whereas coinhibition via PD-1 or BTLA strongly reduced the activation of NF-κB and NFAT. Engagement of BTLA significantly inhibited AP-1, whereas PD-1 had little effect on the activation of this transcription factor. Our triple parameter reporter T cell line is an excellent tool to assess the effect of costimulatory and coinhibitory receptors on NF-κB, NFAT and AP-1 activity and has a wide range of applications beyond the evaluation of costimulatory pathways.
From NLM Medline
- 52Zhang, L.; Morgan, R. A.; Beane, J. D.; Zheng, Z.; Dudley, M. E.; Kassim, S. H.; Nahvi, A. V.; Ngo, L. T.; Sherry, R. M.; Phan, G. Q. Tumor-infiltrating lymphocytes genetically engineered with an inducible gene encoding interleukin-12 for the immunotherapy of metastatic melanoma. Clin. Cancer Res. 2015, 21 (10), 2278– 2288, DOI: 10.1158/1078-0432.CCR-14-208552https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXoslOktbk%253D&md5=7fb2e4d9251f1e3af67d1e0f826a6eefTumor-Infiltrating Lymphocytes Genetically Engineered with an Inducible Gene Encoding Interleukin-12 for the Immunotherapy of Metastatic MelanomaZhang, Ling; Morgan, Richard A.; Beane, Joal D.; Zheng, Zhili; Dudley, Mark E.; Kassim, Sadik H.; Nahvi, Azam V.; Ngo, Lien T.; Sherry, Richard M.; Phan, Giao Q.; Hughes, Marybeth S.; Kammula, Udai S.; Feldman, Steven A.; Toomey, Mary Ann; Kerkar, Sid P.; Restifo, Nicholas P.; Yang, James C.; Rosenberg, Steven A.Clinical Cancer Research (2015), 21 (10), 2278-2288CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: Infusion of interleukin-12 (IL12) can mediate antitumor immunity in animal models, yet its systemic administration to patients with cancer results in minimal efficacy and severe toxicity. Here, we evaluated the antitumor activity of adoptively transferred human tumor-infiltrating lymphocytes (TILs) genetically engineered to secrete single-chain IL12 selectively at the tumor site. Exptl. Design: Thirty-three patients with metastatic melanoma were treated in a cell dose-escalation trial of autologous TILs transduced with a gene encoding a single-chain IL12 driven by a nuclear factor of the activated T cells promoter (NFAT.IL12). No IL2 was administered. Results: The administration of 0.001 to 0.1 × 109 NFAT.IL12-transduced TILs to 17 patients resulted in a single, objective response (5.9%). However, at doses between 0.3 and 3 × 109 cells, 10 of 16 patients (63%) exhibited objective clin. responses. The responses tended to be short, and the administered IL12-producing cells rarely persisted at 1 mo. Increasing cell doses were assocd. with high serum levels of IL12 and IFNγ as well as clin. toxicities, including liver dysfunction, high fevers, and sporadic life-threatening hemodynamic instability. Conclusions: In this first-in-man trial, administration of TILs transduced with an inducible IL12 gene mediated tumor responses in the absence of IL2 administration using cell doses 10- to 100-fold lower than conventional TILs. However, due to toxicities, likely attributable to the secreted IL12, further refinement will be necessary before this approach can be safely used in the treatment of cancer patients.
From NLM Medline
- 53Pegram, H. J.; Lee, J. C.; Hayman, E. G.; Imperato, G. H.; Tedder, T. F.; Sadelain, M.; Brentjens, R. J. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood 2012, 119 (18), 4133– 4141, DOI: 10.1182/blood-2011-12-40004453https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmvV2rsLg%253D&md5=c2c73f661b25d5c17d2f8a529328e4a3Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioningPegram, Hollie J.; Lee, James C.; Hayman, Erik G.; Imperato, Gavin H.; Tedder, Thomas F.; Sadelain, Michel; Brentjens, Renier J.Blood (2012), 119 (18), 4133-4141CODEN: BLOOAW; ISSN:0006-4971. (American Society of Hematology)Adoptive cell therapy with tumor-targeted T cells is a promising approach to cancer therapy. Enhanced clin. outcome using this approach requires conditioning regimens with total body irradn., lymphodepleting chemotherapy, and/or addnl. cytokine support. However, the need for prior conditioning precludes optimal application of this approach to a significant no. of cancer patients intolerant to these regimens. Herein, we present preclin. studies demonstrating that treatment with CD19-specific, chimeric antigen receptor (CAR)-modified T cells that are further modified to constitutively secrete IL-12 are able to safely eradicate established disease in the absence of prior conditioning. We demonstrate in a novel syngeneic tumor model that tumor elimination requires both CD4+ and CD8+ T-cell subsets, autocrine IL-12 stimulation, and subsequent IFNγ secretion by the CAR+ T cells. Importantly, IL-12-secreting, tumor-targeted T cells acquire intrinsic resistance to T regulatory cell-mediated inhibition. Based on these preclin. data, we anticipate that adoptive therapy using CAR-targeted T cells modified to secrete IL-12 will obviate or reduce the need for potentially hazardous conditioning regimens to achieve optimal antitumor responses in cancer patients.
From NLM Medline
- 54Gioux, S.; Choi, H. S.; Frangioni, J. V. Image-guided surgery using invisible near-infrared light: fundamentals of clinical translation. Mol. Imaging 2010, 9 (5), 237– 255, DOI: 10.2310/7290.2010.0003454https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsVejtLbP&md5=08c56d952550d381f30e3f3f5f4e3b05Image-guided surgery using invisible near-infrared light: fundamentals of clinical translationGioux, Sylvain; Choi, Hak Soo; Frangioni, John V.Molecular Imaging (2010), 9 (5), 237-255CODEN: MIOMBP; ISSN:1535-3508. (BC Decker Inc.)A review. The field of biomedical optics has matured rapidly over the last decade and is poised to make a significant impact on patient care. In particular, wide-field (typically > 5 cm), planar, near-IR (NIR) fluorescence imaging has the potential to revolutionize human surgery by providing real-time image guidance to surgeons for tissue that needs to be resected, such as tumors, and tissue that needs to be avoided, such as blood vessels and nerves. However, to become a clin. reality, optimized imaging systems and NIR fluorescent contrast agents will be needed. In this review, we introduce the principles of NIR fluorescence imaging, analyze existing NIR fluorescence imaging systems, and discuss the key parameters that guide contrast agent development. We also introduce the complexities surrounding clin. translation using our experience with the Fluorescence-Assisted Resection and Exploration (FLARE) imaging system as an example. Finally, we introduce state-of-the-art optical imaging techniques that might someday improve image-guided surgery even further.
- 55Mondal, S. B.; Gao, S.; Zhu, N.; Liang, R.; Gruev, V.; Achilefu, S. Real-time fluorescence image-guided oncologic surgery. Adv. Cancer Res. 2014, 124, 171– 211, DOI: 10.1016/B978-0-12-411638-2.00005-755https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXptlOjsbs%253D&md5=5ab78702b3cd0c27e9e3062c583ece83Real-time fluorescence image-guided oncologic surgeryMondal, Suman B.; Gao, Shengkui; Zhu, Nan; Liang, Rongguang; Gruev, Viktor; Achilefu, SamuelAdvances in Cancer Research (2014), 124 (Emerging Applications of Molecular Imaging to Oncology), 171-211CODEN: ACRSAJ; ISSN:0065-230X. (Elsevier Inc.)Medical imaging plays a crit. role in cancer diagnosis and planning. Many of these patients rely on surgical intervention for curative outcomes. This requires a careful identification of the primary and microscopic tumors, and the complete removal of cancer. Although there have been efforts to adapt traditional-imaging modalities for intraoperative image guidance, they suffer from several constraints such as large hardware footprint, high-operation cost, and disruption of the surgical workflow. Because of the ease of image acquisition, relatively low-cost devices and intuitive operation, optical imaging methods have received tremendous interests for use in real-time image-guided surgery. To improve imaging depth under low interference by tissue autofluorescence, many of these applications utilize light in the near-IR (NIR) wavelengths, which is invisible to human eyes. With the availability of a wide selection of tumor-avid contrast agents, advancements in imaging sensors, electronic and optical designs, surgeons are able to combine different attributes of NIR optical imaging techniques to improve treatment outcomes. The emergence of diverse com. and exptl. image guidance systems, which are in various stages of clin. translation, attests to the potential high impact of intraoperative optical imaging methods to improve speed of oncol. surgery with high accuracy and minimal margin positivity.
- 56Stummer, W.; Pichlmeier, U.; Meinel, T.; Wiestler, O. D.; Zanella, F.; Reulen, H. J.; Group, A. L.-G. S. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 2006, 7 (5), 392– 401, DOI: 10.1016/S1470-2045(06)70665-956https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XjvFWlsb0%253D&md5=5f3656046a38c1f4324e4b7715d3995fFluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomized controlled multicenter phase III trialStummer, Walter; Pichlmeier, Uwe; Meinel, Thomas; Wiestler, Otmar Dieter; Zanella, Friedhelm; Reulen, Hans-Juergen; Oppel, F.; Brune, A.; Lanksch, W.; Woiciechowsky, C.; Brock, M.; Vesper, J.; Tonn, J-C.; Goetz, C.; Mayfrank, L.; Oertel, M. F.; Seifert, V.; Franz, K.; Bink, A.; Schackert, G.; Pinzer, T.; Hassler, W.; Bani, A.; Meisel, H-J.; Kern, B. C.; Mehdorn, H. M.; Nabavi, A.; Brawanski, A.; Ullrich, W. W.; Boeker, D. K.; Winking, M.; Weber, F.; Langenbach, U.; Kaehler, U.; Arnold, H.; Knopp, U.; Grumme, T.; Stretz, T.; Stolke, D.; Wiedemayer, H.; Turowski, B.; Pietsch, T.Lancet Oncology (2006), 7 (5), 392-401CODEN: LOANBN; ISSN:1470-2045. (Elsevier Ltd.)5-Aminolevulinic acid is a non-fluorescent prodrug that leads to intracellular accumulation of fluorescent porphyrins in malignant gliomas-a finding that is under investigation for intraoperative identification and resection of these tumors. We aimed to assess the effect of fluorescence-guided resection with 5-aminolevulinic acid on surgical radicality, progression-free survival, overall survival, and morbidity. Three hundred and twenty-two patients aged 23-73 years with suspected malignant glioma amenable to complete resection of contrast-enhancing tumor were randomly assigned to 20 mg/kg bodyweight 5-aminolevulinic acid for fluorescence-guided resection (n = 161) or to conventional microsurgery with white light (n = 161). The primary endpoints were the no. of patients without contrast-enhancing tumor on early MRI (ie, that obtained within 72 h after surgery) and 6-mo progression-free survival as assessed by MRI. Secondary endpoints were vol. of residual tumor on postoperative MRI, overall survival, neurol. deficit, and toxic effects. We report the results of an interim anal. with 270 patients in the full-anal. population (139 assigned 5-aminolevulinic acid, 131 assigned white light), which excluded patients with ineligible histol. and radiol. findings as assessed by central reviewers who were masked as to treatment allocation; the interim anal. resulted in termination of the study as defined by the protocol. Primary and secondary endpoints were analyzed by intention to treat in the full-anal. population. The study is registered at as NCT00241670. Median follow-up was 35.4 mo (95% CI 1.0-56.7). Contrast-enhancing tumor was reseted completely in 90 (65%) of 139 patients assigned 5-aminolevulinic acid compared with 47 (36%) of 131 assigned white light (difference between groups 29% [95% CI 17-40], p < 0.0001). Patients allocated 5-aminolevulinic acid had higher 6-mo progression free survival than did those allocated white light (41.0% [32.8-49.2] vs. 21.1% [14.0-28.2]; difference between groups 19.9% [9.1-30.7], p = 0.0003, Z test). Groups did not differ in the frequency of severe adverse events or adverse events in any organ system class reported within 7 days after surgery. Tumor fluorescence derived from 5-aminolevulinic acid enables more complete resection of contrast-enhancing tumor, leading to improved progression-free survival in patients with malignant glioma.
- 57Bardhan, A.; Deiters, A. Development of photolabile protecting groups and their application to the optochemical control of cell signaling. Curr. Opin. Struct. Biol. 2019, 57, 164– 175, DOI: 10.1016/j.sbi.2019.03.02857https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXnvV2nurY%253D&md5=45fe067abf5f661ff7e572a31da408a8Development of photolabile protecting groups and their application to the optochemical control of cell signalingBardhan, Anirban; Deiters, AlexanderCurrent Opinion in Structural Biology (2019), 57 (), 164-175CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)Many biol. processes are naturally regulated with spatiotemporal control. In order to perturb and investigate them, optochem. tools have been developed that convey similar spatiotemporal precision. Pivotal to optochem. probes are photolabile protecting groups, so called caging groups, and recent developments have enabled new applications to cellular processes, including cell signaling. This review focuses on the advances made in the field of caging groups and their application in cell signaling through caged mols. such as neurotransmitters, lipids, secondary messengers, and proteins.
From NLM Medline
- 58Di Stasi, A.; Tey, S. K.; Dotti, G.; Fujita, Y.; Kennedy-Nasser, A.; Martinez, C.; Straathof, K.; Liu, E.; Durett, A. G.; Grilley, B. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 2011, 365 (18), 1673– 1683, DOI: 10.1056/NEJMoa110615258https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsVWqtbbF&md5=5adcc0e5acc341b06e4e9472959961f0Inducible apoptosis as a safety switch for adoptive cell therapyDi Stasi, Antonio; Tey, Siok-Keen; Dotti, Gianpietro; Fujita, Yuriko; Kennedy-Nasser, Alana; Martinez, Caridad; Straathof, Karin; Liu, Enli; Durett, April G.; Grilley, Bambi; Liu, Hao; Cruz, Conrad R.; Savoldo, Barbara; Gee, Adrian P.; Schindler, John; Krance, Robert A.; Heslop, Helen E.; Spencer, David M.; Rooney, Cliona M.; Brenner, Malcolm K.New England Journal of Medicine (2011), 365 (18), 1673-1683CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)A review. BACKGROUND Cellular therapies could play a role in cancer treatment and regenerative medicine if it were possible to quickly eliminate the infused cells in case of adverse events. We devised an inducible T-cell safety switch that is based on the fusion of human caspase 9 to a modified human FK-binding protein, allowing conditional dimerization. When exposed to a synthetic dimerizing drug, the inducible caspase 9 (iCasp9) becomes activated and leads to the rapid death of cells expressing this construct. METHODS We tested the activity of our safety switch by introducing the gene into donor T cells given to enhance immune reconstitution in recipients of haploidentical stem-cell transplants. Patients received AP1903, an otherwise bioinert small-mol. dimerizing drug, if graft-vs.-host disease (GVHD) developed. We measured the effects of AP1903 on GVHD and on the function and persistence of the cells contg. the iCasp9 safety switch. RESULTS Five patients between the ages of 3 and 17 years who had undergone stem-cell transplantation for relapsed acute leukemia were treated with the genetically modified T cells. The cells were detected in peripheral blood from all five patients and increased in no. over time, despite their constitutive transgene expression. A single dose of dimerizing drug, given to four patients in whom GVHD developed, eliminated more than 90% of the modified T cells within 30 min after administration and ended the GVHD without recurrence. CONCLUSIONS The iCasp9 cell-suicide system may increase the safety of cellular therapies and expand their clin. applications.
- 59Wu, C. Y.; Roybal, K. T.; Puchner, E. M.; Onuffer, J.; Lim, W. A. Remote control of therapeutic T cells through a small molecule-gated chimeric receptor. Science 2015, 350 (6258), aab4077, DOI: 10.1126/science.aab4077There is no corresponding record for this reference.
- 60Reczek, C. R.; Chandel, N. S. The Two Faces of Reactive Oxygen Species in Cancer. Ann. Rev. Cancer Bio. 2017, 1 (1), 79– 98, DOI: 10.1146/annurev-cancerbio-041916-065808There is no corresponding record for this reference.
- 61Sharma, A.; Arambula, J. F.; Koo, S.; Kumar, R.; Singh, H.; Sessler, J. L.; Kim, J. S. Hypoxia-targeted drug delivery. Chem. Soc. Rev. 2019, 48 (3), 771– 813, DOI: 10.1039/C8CS00304A61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFOms7nK&md5=37bbc627b91a138d5e8fdef25869dd29Hypoxia-targeted drug deliverySharma, Amit; Arambula, Jonathan F.; Koo, Seyoung; Kumar, Rajesh; Singh, Hardev; Sessler, Jonathan L.; Kim, Jong SeungChemical Society Reviews (2019), 48 (3), 771-813CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. Hypoxia is a state of low oxygen tension found in numerous solid tumors. It is typically assocd. with abnormal vasculature, which results in a reduced supply of oxygen and nutrients, as well as impaired delivery of drugs. The hypoxic nature of tumors often leads to the development of localized heterogeneous environments characterized by variable oxygen concns., relatively low pH, and increased levels of reactive oxygen species (ROS). The hypoxic heterogeneity promotes tumor invasiveness, metastasis, angiogenesis, and an increase in multidrug-resistant proteins. These factors decrease the therapeutic efficacy of anticancer drugs and can provide a barrier to advancing drug leads beyond the early stages of preclin. development. This review highlights various hypoxia-targeted and activated design strategies for the formulation of drugs or prodrugs and their mechanism of action for tumor diagnosis and treatment.
- 62Wang, M.; Zhao, J.; Zhang, L.; Wei, F.; Lian, Y.; Wu, Y.; Gong, Z.; Zhang, S.; Zhou, J.; Cao, K. Role of tumor microenvironment in tumorigenesis. J. Cancer 2017, 8 (5), 761– 773, DOI: 10.7150/jca.1764862https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVCrsLnI&md5=1485e1cc6546c11fa3bcf6ab72c82b7bRole of tumor microenvironment in tumorigenesisWang, Maonan; Zhao, Jingzhou; Zhang, Lishen; Wei, Fang; Lian, Yu; Wu, Yingfeng; Gong, Zhaojian; Zhang, Shanshan; Zhou, Jianda; Cao, Ke; Li, Xiayu; Xiong, Wei; Li, Guiyuan; Zeng, Zhaoyang; Guo, CanJournal of Cancer (Wyoming, Australia) (2017), 8 (5), 761-773CODEN: JCWAAL; ISSN:1837-9664. (Ivyspring International Publisher Pty Ltd.)Tumorigenesis is a complex and dynamic process, consisting of three stages: initiation, progression, and metastasis. Tumors are encircled by extracellular matrix (ECM) and stromal cells, and the physiol. state of the tumor microenvironment (TME) is closely connected to every step of tumorigenesis. Evidence suggests that the vital components of the TME are fibroblasts and myofibroblasts, neuroendocrine cells, adipose cells, immune and inflammatory cells, the blood and lymphatic vascular networks, and ECM. This manuscript, based on the current studies of the TME, offers a more comprehensive overview of the primary functions of each component of the TME in cancer initiation, progression, and invasion. The manuscript also includes primary therapeutic targeting markers for each player, which may be helpful in treating tumors.
- 63Boedtkjer, E.; Pedersen, S. F. The Acidic Tumor Microenvironment as a Driver of Cancer. Annu. Rev. Physiol. 2020, 82 (1), 103– 126, DOI: 10.1146/annurev-physiol-021119-03462763https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitFKitbjL&md5=72d46086784f0c569ef1d1086087af73The Acidic Tumor Microenvironment as a Driver of CancerBoedtkjer, Ebbe; Pedersen, Stine F.Annual Review of Physiology (2020), 82 (), 103-126CODEN: ARPHAD; ISSN:0066-4278. (Annual Reviews)A review. Acidic metabolic waste products accumulate in the tumor microenvironment because of high metabolic activity and insufficient perfusion. In tumors, the acidity of the interstitial space and the relatively well-maintained intracellular pH influence cancer and stromal cell function, their mutual interplay, and their interactions with the extracellular matrix. Tumor pH is spatially and temporally heterogeneous, and the fitness advantage of cancer cells adapted to extracellular acidity is likely particularly evident when they encounter less acidic tumor regions, for instance, during invasion. Through complex effects on genetic stability, epigenetics, cellular metab., proliferation, and survival, the compartmentalized pH microenvironment favors cancer development. Cellular selection exacerbates the malignant phenotype, which is further enhanced by acid-induced cell motility, extracellular matrix degrdn., attenuated immune responses, and modified cellular and intercellular signaling. In this review, we discuss how the acidity of the tumor microenvironment influences each stage in cancer development, from dysplasia to full-blown metastatic disease.
- 64Kochenderfer, J. N.; Feldman, S. A.; Zhao, Y.; Xu, H.; Black, M. A.; Morgan, R. A.; Wilson, W. H.; Rosenberg, S. A. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J. Immunother. 2009, 32 (7), 689– 702, DOI: 10.1097/CJI.0b013e3181ac613864https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXovFSisrs%253D&md5=422c0e7057c58dffbe5f4ab46e9f4b46Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptorKochenderfer, James N.; Feldman, Steven A.; Zhao, Yangbing; Xu, Hui; Black, Mary A.; Morgan, Richard A.; Wilson, Wyndham H.; Rosenberg, Steven A.Journal of Immunotherapy (2009), 32 (7), 689-702CODEN: JOIMF8; ISSN:1524-9557. (Lippincott Williams & Wilkins)T cells can be engineered to express the genes of chimeric antigen receptors (CARs) that recognize tumor-assocd. antigens. We constructed and compared 2 CARs that contained a single chain variable region moiety that recognized CD19. One CAR contained the signaling moiety of the 4-1BB mol. and the other did not. We selected the CAR that did not contain the 4-1BB moiety for further preclin. development. We demonstrated that gammaretroviruses encoding this receptor could transduce human T cells. Anti-CD19-CAR-transduced CD8 and CD4 T cells produced interferon-γ and interleukin-2 specifically in response to CD19 target cells. The transduced T cells specifically killed primary chronic lymphocytic leukemia (CLL) cells. We transduced T cells from CLL patients that had been previously treated with chemotherapy. We induced these T cells to proliferate sufficiently to provide enough cells for clin. adoptive T cell transfer with a protocol consisting of an initial stimulation with an anti-CD3 monoclonal antibody (OKT3) before transduction followed by a second OKT3 stimulation 7 days after transduction. This protocol was successfully adapted for use in CLL patients with high peripheral blood leukemia cell counts by depleting CD19 cells before the initial OKT3 stimulation. In prepn. for a clin. trial that will enroll patients with advanced B cell malignancies, we generated a producer cell clone that produces retroviruses encoding the anti-CD19 CAR, and we produced sufficient retroviral supernatant for the proposed clin. trial under good manufg. practice conditions.
From NLM Medline
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acssynbio.3c00320.
Supplementary figures showing chemical synthesis of the SMcl OFF-switch adaptor; adaptor biotin quantification; mSA2 CAR expression; T cell subset analysis; functional data for rituximab adaptors; cell viability following UV exposure; and adaptor combination lysis control experiments and supplementary methods describing chemical synthesis of the SMcl OFF-switch adaptor and supplementary references (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.