51
|
Santich BH, Cheung NKV, Klein C. Editorial: Bispecific Antibodies for T-Cell Based Immunotherapy. Front Oncol 2020; 10:628005. [PMID: 33363043 PMCID: PMC7756084 DOI: 10.3389/fonc.2020.628005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/16/2020] [Indexed: 01/14/2023] Open
Affiliation(s)
- Brian H Santich
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Christian Klein
- Cancer Immunotherapy Discovery, Roche Innovation Center Zurich, Schlieren, Switzerland
| |
Collapse
|
52
|
Voynov V, Adam PJ, Nixon AE, Scheer JM. Discovery Strategies to Maximize the Clinical Potential of T-Cell Engaging Antibodies for the Treatment of Solid Tumors. Antibodies (Basel) 2020; 9:E65. [PMID: 33217946 PMCID: PMC7709135 DOI: 10.3390/antib9040065] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/21/2020] [Accepted: 11/11/2020] [Indexed: 12/28/2022] Open
Abstract
T-cell Engaging bispecific antibodies (TcEs) that can re-direct cytotoxic T-cells to kill cancer cells have been validated in clinical studies. To date, the clinical success with these agents has mainly been seen in hematologic tumor indications. However, an increasing number of TcEs are currently being developed to exploit the potent mode-of-action to treat solid tumor indications, which is more challenging in terms of tumor-cell accessibility and the complexity of the tumor microenvironment (TME). Of particular interest is the potential of TcEs as an immunotherapeutic approach for the treatment of non-immunogenic (often referred to as cold) tumors that do not respond to checkpoint inhibitors such as programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) antibodies. This has led to considerable discovery efforts for, firstly, the identification of tumor selective targeting approaches that can safely re-direct cytotoxic T-cells to cancer cells, and, secondly, bispecific antibodies and their derivatives with drug-like properties that promote a potent cytolytic synapse between T-cells and tumor cells, and in the most advanced TcEs, have IgG-like pharmacokinetics for dosing convenience. Based on encouraging pre-clinical data, a growing number of TcEs against a broad range of targets, and using an array of different molecular structures have entered clinical studies for solid tumor indications, and the first clinical data is beginning to emerge. This review outlines the different approaches that have been taken to date in addressing the challenges of exploiting the TcE mode-of-action for a broad range of solid indications, as well as opportunities for future discovery potential.
Collapse
Affiliation(s)
- Vladimir Voynov
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, USA; (A.E.N.); (J.M.S.)
| | - Paul J. Adam
- Cancer Immunology & Immune Modulation, Boehringer Ingelheim RCV GmbH & Co KG, Dr. Boehringer-Gasse 5-11, 1121 Vienna, Austria;
| | - Andrew E. Nixon
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, USA; (A.E.N.); (J.M.S.)
| | - Justin M. Scheer
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, USA; (A.E.N.); (J.M.S.)
| |
Collapse
|
53
|
Li L, Deng L, Meng X, Gu C, Meng L, Li K, Zhang X, Meng Y, Xu W, Zhao L, Chen J, Zhu Z, Huang H. Tumor-targeting anti-EGFR x anti-PD1 bispecific antibody inhibits EGFR-overexpressing tumor growth by combining EGFR blockade and immune activation with direct tumor cell killing. Transl Oncol 2020; 14:100916. [PMID: 33129108 PMCID: PMC7585148 DOI: 10.1016/j.tranon.2020.100916] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/19/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
The anti-PD1 x anti-EGFR bispecific antibody (BsAb) exhibited all in-vitro bioactivities comparable to that of the parental mAbs and showed anti-tumor efficacies of each of the two arms on par with the mAbs in-vivo. The anti-PD1 x anti-EGFR bispecific antibody (BsAb) retained full ADCC towards cancer cells but not to T cells. Thus the BsAb is capable of killing tumor cells via ADCC while sparing T cells for T cell-induced anti-tumor immunity. The anti-PD1 x anti-EGFR bispecific antibody (BsAb) exhibited significantly stronger tumor cell killing effects in the presence of PBMC relative to that of combination of cetuximab with an anti-PD1 mAb, 609A.
We developed a strategy to combine conventional targeted therapy with immune checkpoint blockade using a tumor-targeting bispecific antibody (BsAb) to treat solid tumors. The BsAb was designed to simultaneously engage a tumor-associated antigen, epidermal growth factor receptor (EGFR), and programed cell death protein 1 (PD1). In addition to its direct anti-tumor activity via EGFR inhibition, the BsAb mediated efficient antibody-dependent cellular cytotoxicity (ADCC) and activated T cell antitumor im munity through blockade of PD1 from interacting with its counterpart, programed cell death ligand 1 (PDL1). Further, the BsAb exhibited a potent direct tumor cell killing activity in the presence of PBMC, most likely, via activating and, at the same time, physically engaging T cells with tumor cells. Taken together, we here illustrate a new strategy in the design and production of novel BsAbs with enhanced therapeutic efficacy through both direct tumor growth inhibition and T cell activation via tumor-targeted immune checkpoint blockade.
Collapse
Affiliation(s)
- Li Li
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Lan Deng
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Xiaoqing Meng
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Changling Gu
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Li Meng
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Kai Li
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Xuesai Zhang
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Yun Meng
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Wei Xu
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Le Zhao
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Jianhe Chen
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Zhenping Zhu
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China.
| | - Haomin Huang
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China.
| |
Collapse
|
54
|
Groeneveldt C, Kinderman P, van den Wollenberg DJM, van den Oever RL, Middelburg J, Mustafa DAM, Hoeben RC, van der Burg SH, van Hall T, van Montfoort N. Preconditioning of the tumor microenvironment with oncolytic reovirus converts CD3-bispecific antibody treatment into effective immunotherapy. J Immunother Cancer 2020; 8:jitc-2020-001191. [PMID: 33082167 PMCID: PMC7577070 DOI: 10.1136/jitc-2020-001191] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2020] [Indexed: 12/14/2022] Open
Abstract
Background T-cell-engaging CD3-bispecific antibodies (CD3-bsAbs) are promising modalities for cancer immunotherapy. Although this therapy has reached clinical practice for hematological malignancies, the absence of sufficient infiltrating T cells is a major barrier for efficacy in solid tumors. In this study, we exploited oncolytic reovirus as a strategy to enhance the efficacy of CD3-bsAbs in immune-silent solid tumors. Methods The mutant p53 and K-ras induced murine pancreatic cancer model KPC3 resembles human pancreatic ductal adenocarcinomas with a desmoplastic tumor microenvironment, low T-cell density and resistance to immunotherapy. Immune-competent KPC3 tumor-bearing mice were intratumorally injected with reovirus type 3 Dearing strain and the reovirus-induced changes in the tumor microenvironment and spleen were analyzed over time by NanoString analysis, quantitative RT-PCR and multicolor flow cytometry. The efficacy of reovirus in combination with systemically injected CD3-bsAbs was evaluated in immune-competent mice with established KPC3 or B16.F10 tumors, and in the close-to-patient human epidermal growth factor receptor 2 (HER2)+ breast cancer model BT474 engrafted in immunocompromised mice with human T cells as effector cells. Results Replication-competent reovirus induced an early interferon signature, followed by a strong influx of natural killer cells and CD8+ T cells, at the cost of FoxP3+ Tregs. Viral replication declined after 7 days and was associated with a systemic activation of lymphocytes and the emergence of intratumoral reovirus-specific CD8+ T cells. Although tumor-infiltrating T cells were mostly reovirus-specific and not tumor-specific, they served as non-exhausted effector cells for the subsequently systemically administered CD3-bsAbs. Combination treatment of reovirus and CD3-bsAbs led to the regression of large, established KPC3, B16.F10 and BT474 tumors. Reovirus as a preconditioning regimen performed significantly better than simultaneous or early administration of CD3-bsAbs. This combination treatment induced regressions of distant lesions that were not injected with reovirus, and systemic administration of both reovirus and CD3-bsAbs also led to tumor control. This suggests that this therapy might also be effective for metastatic disease. Conclusions Oncolytic reovirus administration represents an effective strategy to induce a local interferon response and strong T-cell influx, thereby sensitizing the tumor microenvironment for subsequent CD3-bsAb therapy. This combination therapy warrants further investigation in patients with non-inflamed solid tumors.
Collapse
Affiliation(s)
- Christianne Groeneveldt
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Priscilla Kinderman
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ruben L van den Oever
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Dana A M Mustafa
- Department of Pathology, Tumor Immuno-Pathology Laboratory, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rob C Hoeben
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Nadine van Montfoort
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
55
|
Guo ZS, Lotze MT, Zhu Z, Storkus WJ, Song XT. Bi- and Tri-Specific T Cell Engager-Armed Oncolytic Viruses: Next-Generation Cancer Immunotherapy. Biomedicines 2020; 8:E204. [PMID: 32664210 PMCID: PMC7400484 DOI: 10.3390/biomedicines8070204] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses (OVs) are potent anti-cancer biologics with a bright future, having substantial evidence of efficacy in patients with cancer. Bi- and tri-specific antibodies targeting tumor antigens and capable of activating T cell receptor signaling have also shown great promise in cancer immunotherapy. In a cutting-edge strategy, investigators have incorporated the two independent anti-cancer modalities, transforming them into bi- or tri-specific T cell engager (BiTE or TriTE)-armed OVs for targeted immunotherapy. Since 2014, multiple research teams have studied this combinatorial strategy, and it showed substantial efficacy in various tumor models. Here, we first provide a brief overview of the current status of oncolytic virotherapy and the use of multi-specific antibodies for cancer immunotherapy. We then summarize progress on BiTE and TriTE antibodies as a novel class of cancer therapeutics in preclinical and clinical studies, followed by a discussion of BiTE- or TriTE-armed OVs for cancer therapy in translational models. In addition, T cell receptor mimics (TCRm) have been developed into BiTEs and are expected to greatly expand the application of BiTEs and BiTE-armed OVs for the effective targeting of intracellular tumor antigens. Future applications of such innovative combination strategies are emerging as precision cancer immunotherapies.
Collapse
Affiliation(s)
- Zong Sheng Guo
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael T Lotze
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Zhi Zhu
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Walter J Storkus
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
56
|
Chornoguz O, Leettola CN, Leander K, Brosnan K, Emmell E, Chiu ML, Santulli-Marotto S. Characterization of a Novel Bispecific Antibody That Activates T Cells In Vitro and Slows Tumor Growth In Vivo. Monoclon Antib Immunodiagn Immunother 2020; 38:242-254. [PMID: 31825302 PMCID: PMC6918852 DOI: 10.1089/mab.2019.0035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Although CD3 T cell redirecting antibodies have been successfully utilized for the treatment of hematological malignancies (blinatumomab), the T cell signaling pathways induced by these molecules are incompletely understood. To gain insight into the mechanism of action for T cell redirection antibodies, we created a novel murine CD3xEpCAM bispecific antibody that incorporates a silent Fc to dissect function and signaling of murine CD8 OT1 T cells upon stimulation. T cell-mediated cytotoxicity, cytokine secretion, expression of activation markers, and proliferation were directly induced in T cells treated with the novel CD3xEpCAM bispecific molecule in vitro in the presence of epithelial cell adhesion molecule (EpCAM) expressing tumor cells. Nanostring analysis showed that CD3xEpCAM induced a gene expression profile that resembled antigen-mediated activation, although the magnitude was lower than that of the antigen-induced response. In addition, this CD3xEpCAM bispecific antibody exhibited in vivo efficacy. This is the first study that investigates both in vitro and in vivo murine CD8 T cell function and signaling induced by a CD3xEpCAM antibody having a silent Fc to delineate differences between antigen-independent and antigen-specific T cell activation. These findings expand the understanding of T cell function and signaling induced by CD3 redirection bispecific antibodies and may help to develop more efficacious CD3 redirection therapeutics for cancer treatment, particularly for solid tumors.
Collapse
Affiliation(s)
- Olesya Chornoguz
- Janssen Biotherapeutics, Janssen R&D, Spring House, Pennsylvania
| | | | - Karen Leander
- Janssen Biotherapeutics, Janssen R&D, Spring House, Pennsylvania
| | - Kerry Brosnan
- Janssen Biotherapeutics, Janssen R&D, Spring House, Pennsylvania
| | - Eva Emmell
- Janssen Biotherapeutics, Janssen R&D, Spring House, Pennsylvania
| | - Mark L Chiu
- Janssen Biotherapeutics, Janssen R&D, Spring House, Pennsylvania
| | | |
Collapse
|
57
|
Abstract
A bispecific antibody (bsAb) can simultaneously bind two different epitopes or antigens, allowing for multiple mechanistic functions with synergistic effects. BsAbs have attracted significant scientific attentions and efforts towards their development as drugs for cancers. There are 21 bsAbs currently undergoing clinical trials in China. Here, we review their platform technologies, expression and production, and biological activities and bioassay of these bsAbs, and summarize their structural formats and mechanisms of actions. T-cell redirection and checkpoint inhibition are two main mechanisms of the bsAbs that we discuss in detail. Furthermore, we provide our perspective on the future of bsAb development in China, including CD3-bsAbs for solid tumors and related cytokine release syndromes, expression and chemistry, manufacturing and controls, clinical development, and immunogenicity.
Collapse
Affiliation(s)
- Jing Zhang
- Wuhan YZY Biopharma Co., Ltd, Biolake City C2-1, No. 666 Gaoxin Road, Wuhan, Hubei 430075, China
| | - Jizu Yi
- Wuhan YZY Biopharma Co., Ltd, Biolake City C2-1, No. 666 Gaoxin Road, Wuhan, Hubei 430075, China
| | - Pengfei Zhou
- Wuhan YZY Biopharma Co., Ltd, Biolake City C2-1, No. 666 Gaoxin Road, Wuhan, Hubei 430075, China
| |
Collapse
|
58
|
Sow HS, Benonisson H, Brouwers C, Linssen MM, Camps M, Breukel C, Claassens J, van Hall T, Ossendorp F, Fransen MF, Verbeek JS. Immunogenicity of rat-neu + mouse mammary tumours determines the T cell-dependent therapeutic efficacy of anti-neu monoclonal antibody treatment. Sci Rep 2020; 10:3933. [PMID: 32127568 PMCID: PMC7054273 DOI: 10.1038/s41598-020-60893-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 02/14/2020] [Indexed: 11/09/2022] Open
Abstract
The use of Trastuzumab (Herceptin), a monoclonal antibody (mAb) targeting HER2/neu, results in an increased median survival in Her2+ breast cancer patients. The tumour mutational burden and the presence of tumour infiltrating lymphocytes (TILs) clearly correlate with response to trastuzumab. Here, we investigated if the immunogenicity of the transplantable rat-neu+ tumour cell line (TUBO) derived from a BALB/c-NeuT primary tumour is associated with the response to anti-neu mAb therapy. We compared the TUBO tumour outgrowth and tumour infiltrating T cells in isogenic (BALB/c-NeuT) and non-isogenic (WT BALB/c) recipient mice. Furthermore, therapeutic efficacy of anti-neu mAb and the contribution of T cells were examined in both mouse strains. The outgrowth of untreated tumours was significantly better in BALB/c-NeuT than WT BALB/c mice. Moreover, tumour infiltrating T cells were more abundantly present in WT BALB/c than BALB/c-NeuT mice, showing that the TUBO tumour was more immunogenic in WT BALB/c mice. In TUBO tumour bearing WT BALB/c mice, anti-neu mAb therapy resulted in an increase of tumour infiltrating T cells and long-term survival. When T cells were depleted, this strong anti-tumour effect was reduced to an outgrowth delay. In contrast, in TUBO tumour bearing BALB/c-NeuT mice, treatment with anti-neu mAb resulted only in tumour outgrowth delay, both in the presence and absence of T cells. We concluded that in immunogenic tumours the response to anti-neu mAb therapy is enhanced by additional T cell involvement compared to the response to anti-neu mAb in non-immunogenic tumours.
Collapse
Affiliation(s)
- Heng Sheng Sow
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Hreinn Benonisson
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Conny Brouwers
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Margot M Linssen
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marcel Camps
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands
| | - Cor Breukel
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jill Claassens
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands.,Department of Pulmonary Diseases, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands.
| |
Collapse
|
59
|
Liu D. CAR-T "the living drugs", immune checkpoint inhibitors, and precision medicine: a new era of cancer therapy. J Hematol Oncol 2019; 12:113. [PMID: 31703740 PMCID: PMC6842223 DOI: 10.1186/s13045-019-0819-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
New advances in the design and manufacture of monoclonal antibodies, bispecific T cell engagers, and antibody-drug conjugates make the antibody-directed agents more powerful with less toxicities. Small molecule inhibitors are routinely used now as oral targeted agents for multiple cancers. The discoveries of PD1 and PD-L1 as negative immune checkpoints for T cells have led to the revolution of modern cancer immunotherapy. Multiple agents targeting PD1, PD-L1, or CTLA-4 are widely applied as immune checkpoint inhibitors (ICIs) which alleviate the suppression of immune regulatory machineries and lead to immunoablation of once highly refractory cancers such as stage IV lung cancer. Tisagenlecleucel and axicabtagene ciloleucel are the two approved CD19-targeted chimeric antigen receptor (CAR) T cell products. Several CAR-T cell platforms targeting B cell maturation antigen (BCMA) are under active clinical trials for refractory and/or relapsed multiple myeloma. Still more targets such as CLL-1, EGFR, NKG2D and mesothelin are being directed in CAR-T cell trials for leukemia and solid tumors. Increasing numbers of novel agents are being studied to target cancer-intrinsic oncogenic pathways as well as immune checkpoints. One such an example is targeting CD47 on macrophages which represents a "do-not-eat-me" immune checkpoint. Fueling the current excitement of cancer medicine includes also TCR- T cells, TCR-like antibodies, cancer vaccines and oncolytic viruses.
Collapse
Affiliation(s)
- Delong Liu
- New York Medical College, Valhalla, NY, 10595, USA.
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
60
|
Labrijn AF, Janmaat ML, Reichert JM, Parren PWHI. Bispecific antibodies: a mechanistic review of the pipeline. Nat Rev Drug Discov 2019; 18:585-608. [DOI: 10.1038/s41573-019-0028-1] [Citation(s) in RCA: 493] [Impact Index Per Article: 82.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|