1
|
Liu X, Tang G, Chen Y, Li Y, Li H, Wang X. SpatialDeX Is a Reference-Free Method for Cell-Type Deconvolution of Spatial Transcriptomics Data in Solid Tumors. Cancer Res 2025; 85:171-182. [PMID: 39387817 DOI: 10.1158/0008-5472.can-24-1472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/06/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
The rapid development of spatial transcriptomics (ST) technologies has enabled transcriptome-wide profiling of gene expression in tissue sections. Despite the emergence of single-cell resolution platforms, most ST sequencing studies still operate at a multicell resolution. Consequently, deconvolution of cell identities within the spatial spots has become imperative for characterizing cell-type-specific spatial organization. To this end, we developed Spatial Deconvolution Explorer (SpatialDeX), a regression model-based method for estimating cell-type proportions in tumor ST spots. SpatialDeX exhibited comparable performance to reference-based methods and outperformed other reference-free methods with simulated ST data. Using experimental ST data, SpatialDeX demonstrated superior performance compared with both reference-based and reference-free approaches. Additionally, a pan-cancer clustering analysis on tumor spots identified by SpatialDeX unveiled distinct tumor progression mechanisms both within and across diverse cancer types. Overall, SpatialDeX is a valuable tool for unraveling the spatial cellular organization of tissues from ST data without requiring single-cell RNA-seq references. Significance: The development of a reference-free method for deconvolving the identity of cells in spatial transcriptomics datasets enables exploration of tumor architecture to gain deeper insights into the dynamics of the tumor microenvironment.
Collapse
Affiliation(s)
- Xinyi Liu
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Gongyu Tang
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri
| | - Yuhao Chen
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Yuanxiang Li
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Hua Li
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Xiaowei Wang
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois
- University of Illinois Cancer Center, Chicago, Illinois
| |
Collapse
|
2
|
Fattahi AS, Jafari M, Farahavar G, Abolmaali SS, Tamaddon AM. Expanding horizons in cancer therapy by immunoconjugates targeting tumor microenvironments. Crit Rev Oncol Hematol 2024; 201:104437. [PMID: 38977144 DOI: 10.1016/j.critrevonc.2024.104437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
Immunoconjugates are promising molecules combining antibodies with different agents, such as toxins, drugs, radionuclides, or cytokines that primarily aim to target tumor cells. However, tumor microenvironment (TME), which comprises a complex network of various cells and molecular cues guiding tumor growth and progression, remains a major challenge for effective cancer therapy. Our review underscores the pivotal role of TME in cancer therapy with immunoconjugates, examining the intricate interactions with TME and recent advancements in TME-targeted immunoconjugates. We explore strategies for targeting TME components, utilizing diverse antibodies such as neutralizing, immunomodulatory, immune checkpoint inhibitors, immunostimulatory, and bispecific antibodies. Additionally, we discuss different immunoconjugates, elucidating their mechanisms of action, advantages, limitations, and applications in cancer immunotherapy. Furthermore, we highlight emerging technologies enhancing the safety and efficacy of immunoconjugates, such as antibody engineering, combination therapies, and nanotechnology. Finally, we summarize current advancements, perspectives, and future developments of TME-targeted immunoconjugates.
Collapse
Affiliation(s)
- Amir Saamaan Fattahi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mahboobeh Jafari
- Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Ghazal Farahavar
- Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| |
Collapse
|
3
|
Li T, Wang X, Niu M, Wang M, Zhou J, Wu K, Yi M. Bispecific antibody targeting TGF-β and PD-L1 for synergistic cancer immunotherapy. Front Immunol 2023; 14:1196970. [PMID: 37520520 PMCID: PMC10373067 DOI: 10.3389/fimmu.2023.1196970] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
The PD-1/PD-L1 signaling pathway plays a crucial role in cancer immune evasion, and the use of anti-PD-1/PD-L1 antibodies represents a significant milestone in cancer immunotherapy. However, the low response rate observed in unselected patients and the development of therapeutic resistance remain major obstacles to their clinical application. Accumulating studies showed that overexpressed TGF-β is another immunosuppressive factor apart from traditional immune checkpoints. Actually, the effects of PD-1 and TGF-β pathways are independent and interactive, which work together contributing to the immune evasion of cancer cell. It has been verified that blocking TGF-β and PD-L1 simultaneously could enhance the efficacy of PD-L1 monoclonal antibody and overcome its treatment resistance. Based on the bispecific antibody or fusion protein technology, multiple bispecific and bifunctional antibodies have been developed. In the preclinical and clinical studies, these updated antibodies exhibited potent anti-tumor activity, superior to anti-PD-1/PD-L1 monotherapies. In the review, we summarized the advances of bispecific antibodies targeting TGF-β and PD-L1 in cancer immunotherapy. We believe these next-generation immune checkpoint inhibitors would substantially alter the cancer treatment paradigm, especially in anti-PD-1/PD-L1-resistant patients.
Collapse
Affiliation(s)
- Tianye Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Xinrun Wang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Mengke Niu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Mingli Wang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
4
|
Liguori L, Polcaro G, Nigro A, Conti V, Sellitto C, Perri F, Ottaiano A, Cascella M, Zeppa P, Caputo A, Pepe S, Sabbatino F. Bispecific Antibodies: A Novel Approach for the Treatment of Solid Tumors. Pharmaceutics 2022; 14:2442. [PMID: 36432631 PMCID: PMC9694302 DOI: 10.3390/pharmaceutics14112442] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Advancement in sequencing technologies allows for the identification of molecular pathways involved in tumor progression and treatment resistance. Implementation of novel agents targeting these pathways, defined as targeted therapy, significantly improves the prognosis of cancer patients. Targeted therapy also includes the use of monoclonal antibodies (mAbs). These drugs recognize specific oncogenic proteins expressed in cancer cells. However, as with many other types of targeting agents, mAb-based therapy usually fails in the long-term control of cancer progression due to the development of resistance. In many cases, resistance is caused by the activation of alternative pathways involved in cancer progression and the development of immune evasion mechanisms. To overcome this off-target resistance, bispecific antibodies (bsAbs) were developed to simultaneously target differential oncogenic pathway components, tumor-associated antigens (TAA) and immune regulatory molecules. As a result, in the last few years, several bsAbs have been tested or are being tested in cancer patients. A few of them are currently approved for the treatment of some hematologic malignancies but no bsAbs are approved in solid tumors. In this review, we will provide an overview of the state-of-the-art of bsAbs for the treatment of solid malignancies outlining their classification, design, main technologies utilized for production, mechanisms of action, updated clinical evidence and potential limitations.
Collapse
Affiliation(s)
- Luigi Liguori
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
| | - Giovanna Polcaro
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
| | - Annunziata Nigro
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
| | - Valeria Conti
- Clinical Pharmacology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
| | - Carmine Sellitto
- Clinical Pharmacology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
| | - Francesco Perri
- Medical and Experimental Head and Neck Oncology Unit, INT IRCSS, Foundation “G. Pascale”, 80131 Naples, Italy
| | - Alessandro Ottaiano
- SSD Innovative Therapies for Abdominal Metastases, Abdominal Oncology, INT IRCCS Foundation “G. Pascale”, 80131 Naples, Italy
| | - Marco Cascella
- Unit of Anesthesiology and Pain Therapy, INT IRCCS Foundation “G. Pascale”, 80131 Naples, Italy
| | - Pio Zeppa
- Pathology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
| | - Alessandro Caputo
- Pathology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
| | - Stefano Pepe
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
| |
Collapse
|
5
|
Wu Y, Yi M, Zhu S, Wang H, Wu K. Recent advances and challenges of bispecific antibodies in solid tumors. Exp Hematol Oncol 2021; 10:56. [PMID: 34922633 PMCID: PMC8684149 DOI: 10.1186/s40164-021-00250-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/03/2021] [Indexed: 12/29/2022] Open
Abstract
Cancer immunotherapy has made remarkable progress in the past decade. Bispecific antibodies (BsAbs) have acquired much attention as the next generation strategy of antibody-target cancer immunotherapy, which overwhelmingly focus on T cell recruitment and dual receptors blockade. So far, BsAb drugs have been proved clinically effective and approved for the treatment of hematologic malignancies, but no BsAb have been approved in solid tumors. Numerous designed BsAb drugs for solid tumors are now undergoing evaluation in clinical trials. In this review, we will introduce the formats of bispecific antibodies, and then update the latest preclinical studies and clinical trials in solid tumors of BsAbs targeting EpCAM, CEA, PMSA, ErbB family, and so on. Finally, we discuss the BsAb-related adverse effects and the alternative strategy for future study.
Collapse
Affiliation(s)
- Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuangli Zhu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haiyong Wang
- Beijing Anjianxi Medicinal Technology Co., Ltd., No.2 Cuiwei Road, Haidian District, Beijing, 100036, China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
6
|
Zhao C, Zhang W, Gong G, Xie L, Wang MW, Hu Y. A new approach to produce IgG 4-like bispecific antibodies. Sci Rep 2021; 11:18630. [PMID: 34545109 PMCID: PMC8452627 DOI: 10.1038/s41598-021-97393-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 07/05/2021] [Indexed: 11/12/2022] Open
Abstract
While achieving rapid developments in recent years, bispecific antibodies are still difficult to design and manufacture, due to mispair of both heavy and light chains. Here we report a novel technology to make bispecific molecules. The knob-into-hole method was used to pair two distinct heavy chains as a heterodimer. IgG4 S228P CH1-CL interface was then partially replaced by T-cell receptor α/β constant domain to increase the efficiency of cognate heavy and light chain pairing. Following expression and purification, the bispecific antibody interface exchange was confirmed by Western blotting and LC–MS/MS. To ensure its validity, we combined a monovalent bispecific antibody against PD-1 (sequence from Pembrolizumab) and LAG3 (sequence from Relatlimab). The results showed that the molecule could be assembled correctly at a ratio of 95% in cells. In vitro functional assay demonstrated that the purified bispecific antibody exhibits an enhanced agonist activity compared to that of the parental antibodies. Low immunogenicity was predicted by an open-access software and ADA test.
Collapse
Affiliation(s)
- Caizhi Zhao
- School of Pharmacy, Fudan University, Shanghai, 201203, China.,China State Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Wei Zhang
- China State Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Guihua Gong
- China State Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Liping Xie
- China State Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Ming-Wei Wang
- School of Pharmacy, Fudan University, Shanghai, 201203, China. .,The National Center for Drug Screening, Shanghai, 201203, China.
| | - Youjia Hu
- China State Institute of Pharmaceutical Industry, Shanghai, 201203, China.
| |
Collapse
|
7
|
Tuyishime M, Dashti A, Faircloth K, Jha S, Nordstrom JL, Haynes BF, Silvestri G, Chahroudi A, Margolis DM, Ferrari G. Elimination of SHIV Infected Cells by Combinations of Bispecific HIVxCD3 DART ® Molecules. Front Immunol 2021; 12:710273. [PMID: 34484212 PMCID: PMC8415083 DOI: 10.3389/fimmu.2021.710273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/26/2021] [Indexed: 01/13/2023] Open
Abstract
Bispecific HIVxCD3 DART molecules that co-engage the viral envelope glycoprotein (Env) on HIV-1-infected cells and the CD3 receptor on CD3+ T cells are designed to mediate the cytolysis of HIV-1-infected, Env-expressing cells. Using a novel ex vivo system with cells from rhesus macaques (RMs) infected with a chimeric Simian-Human Immunodeficiency Virus (SHIV) CH505 and maintained on ART, we tested the ability of HIVxCD3 DART molecules to mediate elimination of in vitro-reactivated CD4+ T cells in the absence or presence of autologous CD8+ T cells. HIVxCD3 DART molecules with the anti-HIV-1 Env specificities of A32 or 7B2 (non-neutralizing antibodies) or PGT145 (broadly neutralizing antibody) were evaluated individually or combined. DART molecule-mediated antiviral activity increased significantly in the presence of autologous CD8+ T cells. In this ex vivo system, the PGT145 DART molecule was more active than the 7B2 DART molecule, which was more active than the A32 DART molecule. A triple combination of the DART molecules exceeded the activity of the individual PGT145 DART molecule. Modified quantitative virus outgrowth assays confirmed the ability of the DART molecules to redirect RM CD3+ T cells to eliminate SHIV-infected RM CD4+ T cells as demonstrated by the decreased propagation of in vitro infection by the infected cells pre-incubated with DART molecules in presence of effector CD8+ T cells. While mediating cytotoxic activity, DART molecules did not increase proinflammatory cytokine production. In summary, combination of HIVxCD3 DART molecules that have broadly-neutralizing and non-neutralizing anti-HIV-1 Env specificities can leverage the host immune system for treatment of HIV-1 infection but will require appropriate reactivation of the latent reservoir.
Collapse
Affiliation(s)
- Marina Tuyishime
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Amir Dashti
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Katelyn Faircloth
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Shalini Jha
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | | | - Barton F. Haynes
- Duke Human Vaccine Institute, Durham, NC, United States
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| | - Guido Silvestri
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Ann Chahroudi
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| | - David M. Margolis
- University of North Carolina (UNC) HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Guido Ferrari
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
8
|
Maresca KP, Chen J, Mathur D, Giddabasappa A, Root A, Narula J, King L, Schaer D, Golas J, Kobylarz K, Rosfjord E, Keliher E, Chen L, Ram S, Pickering EH, Hardwick JS, Rejto PA, Hussein A, Ilovich O, Staton K, Wilson I, McCarthy TJ. Preclinical Evaluation of 89Zr-Df-IAB22M2C PET as an Imaging Biomarker for the Development of the GUCY2C-CD3 Bispecific PF-07062119 as a T Cell Engaging Therapy. Mol Imaging Biol 2021; 23:941-951. [PMID: 34143379 PMCID: PMC8578158 DOI: 10.1007/s11307-021-01621-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/19/2021] [Accepted: 06/03/2021] [Indexed: 01/10/2023]
Abstract
Purpose A sensitive and specific imaging biomarker to monitor immune activation and quantify pharmacodynamic responses would be useful for development of immunomodulating anti-cancer agents. PF-07062119 is a T cell engaging bispecific antibody that binds to CD3 and guanylyl cyclase C, a protein that is over-expressed by colorectal cancers. Here, we used 89Zr-Df-IAB22M2C (89Zr-Df-Crefmirlimab), a human CD8-specific minibody to monitor CD8+ T cell infiltration into tumors by positron emission tomography. We investigated the ability of 89Zr-Df-IAB22M2C to track anti-tumor activity induced by PF-07062119 in a human CRC adoptive transfer mouse model (with injected activated/expanded human T cells), as well as the correlation of tumor radiotracer uptake with CD8+ immunohistochemical staining. Procedures NOD SCID gamma mice bearing human CRC LS1034 tumors were treated with four different doses of PF-07062119, or a non-targeted CD3 BsAb control, and imaged with 89Zr-Df-IAB22M2C PET at days 4 and 9. Following PET/CT imaging, mice were euthanized and dissected for ex vivo distribution analysis of 89Zr-Df-IAB22M2C in tissues on days 4 and 9, with additional data collected on day 6 (supplementary). Data were analyzed and reported as standard uptake value and %ID/g for in vivo imaging and ex vivo tissue distribution. In addition, tumor tissues were evaluated by immunohistochemistry for CD8+ T cells. Results The results demonstrated substantial mean uptake of 89Zr-Df-IAB22M2C (%ID/g) in PF-07062119-treated tumors, with significant increases in comparison to non-targeted BsAb-treated controls, as well as PF-07062119 dose-dependent responses over time of treatment. A moderate correlation was observed between tumor tissue radioactivity uptake and CD8+ cell density, demonstrating the value of the imaging agent for non-invasive assessment of intra-tumoral CD8+ T cells and the mechanism of action for PF-07062119. Conclusion Immune-imaging technologies for quantitative cellular measures would be a valuable biomarker in immunotherapeutic clinical development. We demonstrated a qualification of 89Zr-IAB22M2C PET to evaluate PD responses (mice) to a novel immunotherapeutic. Supplementary Information The online version contains supplementary material available at 10.1007/s11307-021-01621-0.
Collapse
Affiliation(s)
- Kevin P Maresca
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA.
| | - Jianqing Chen
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA
| | - Divya Mathur
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA.,Regneron Pharmaceuticals, Tarrytown, NY, USA
| | | | - Adam Root
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA.,Generate Biomedicines, Inc, Cambridge, MA, USA
| | - Jatin Narula
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA
| | - Lindsay King
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA
| | - David Schaer
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA
| | - Jonathan Golas
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA.,Regneron Pharmaceuticals, Tarrytown, NY, USA
| | - Keith Kobylarz
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA
| | - Edward Rosfjord
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA.,Black Diamond Therapeutics, New York, NY, USA
| | - Edmund Keliher
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA
| | - Laigao Chen
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA
| | - Sripad Ram
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA
| | - Eve H Pickering
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA
| | - James S Hardwick
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA
| | - Paul A Rejto
- Worldwide Research, Development & Medicine, Pfizer Inc, New York, USA
| | | | - Ohad Ilovich
- Invicro, A Konica Minolta Company, New Haven, USA
| | - Kevin Staton
- Evergreen Theragnostics, Jersey City, NJ, USA.,Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | |
Collapse
|
9
|
Figueroa-Vazquez V, Ko J, Breunig C, Baumann A, Giesen N, Pálfi A, Müller C, Lutz C, Hechler T, Kulke M, Müller-Tidow C, Krämer A, Goldschmidt H, Pahl A, Raab MS. HDP-101, an Anti-BCMA Antibody-Drug Conjugate, Safely Delivers Amanitin to Induce Cell Death in Proliferating and Resting Multiple Myeloma Cells. Mol Cancer Ther 2020; 20:367-378. [PMID: 33298585 DOI: 10.1158/1535-7163.mct-20-0287] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 09/09/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022]
Abstract
Despite major treatment advances in recent years, patients with multiple myeloma inevitably relapse. The RNA polymerase II complex has been identified as a promising therapeutic target in both proliferating and dormant cancer cells. Alpha-amanitin, a toxin so far without clinical application due to high liver toxicity, specifically inhibits this complex. Here, we describe the development of HDP-101, an anti-B-cell maturation antigen (BCMA) antibody conjugated with an amanitin derivative. HDP-101 displayed high efficacy against both proliferating and resting myeloma cells in vitro, sparing BCMA-negative cells. In subcutaneous and disseminated murine xenograft models, HDP-101 induced tumor regression at low doses, including durable complete remissions after a single intravenous dose. In cynomolgus monkeys, HDP-101 was well tolerated with a promising therapeutic index. In conclusion, HDP-101 safely and selectively delivers amanitin to myeloma cells and provides a novel therapeutic approach to overcome drug resistance in this disease.
Collapse
Affiliation(s)
- Vianihuini Figueroa-Vazquez
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Jonathan Ko
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Anja Baumann
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Nicola Giesen
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Anikó Pálfi
- Heidelberg Pharma Research GmbH, Ladenburg, Germany
| | | | | | | | | | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Hartmut Goldschmidt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center of Tumor Diseases (NCT), Heidelberg, Germany
| | - Andreas Pahl
- Heidelberg Pharma Research GmbH, Ladenburg, Germany.
| | - Marc S Raab
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany. .,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Immunotherapy strategies alternative to current antiretroviral therapies will need to address viral diversity while increasing the immune system's ability to efficiently target the latent virus reservoir. Antibody-based molecules can be designed based on broadly neutralizing and non-neutralizing antibodies that target free virions and infected cells. These multispecific molecules, either by IgG-like or non-IgG-like in structure, aim to target several independent HIV-1 epitopes and/or engage effector cells to eliminate the replicating virus and infected cells. This detailed review is intended to stimulate discussion on future requirements for novel immunotherapeutic molecules. RECENT FINDINGS Bispecific and trispecific antibodies are engineered as a single molecules to target two or more independent epitopes on the HIV-1 envelope (Env). These antibody-based molecules have increased avidity for Env, leading to improved neutralization potency and breadth compared with single parental antibodies. Furthermore, bispecific and trispecific antibodies that engage cellular receptors with one arm of the molecule help concentrate inhibitory molecules to the sites of potential infection and facilitate engagement of immune effector cells and Env-expressing target cells for their elimination. SUMMARY Recently engineered antibody-based molecules of different sizes and structures show promise in vitro or in vivo and are encouraging candidates for HIV treatment.
Collapse
Affiliation(s)
- Marina Tuyishime
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Guido Ferrari
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
11
|
Sircar A, Chowdhury SM, Hart A, Bell WC, Singh S, Sehgal L, Epperla N. Impact and Intricacies of Bone Marrow Microenvironment in B-cell Lymphomas: From Biology to Therapy. Int J Mol Sci 2020; 21:E904. [PMID: 32019190 PMCID: PMC7043222 DOI: 10.3390/ijms21030904] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/26/2020] [Accepted: 01/29/2020] [Indexed: 12/14/2022] Open
Abstract
Lymphoma, a group of widely prevalent hematological malignancies of lymphocyte origin, has become the focus of significant clinical research due to their high propensity for refractory/relapsed (R/R) disease, leading to poor prognostic outcomes. The complex molecular circuitry in lymphomas, especially in the aggressive phenotypes, has made it difficult to find a therapeutic option that can salvage R/R disease. Furthermore, the association of lymphomas with the Bone Marrow (BM) microenvironment has been found to portend worse outcomes in terms of heightened chances of relapse and acquired resistance to chemotherapy. This review assesses the current therapy options in three distinct types of lymphomas: diffuse large B-cell lymphoma, follicular lymphoma and mantle cell lymphoma. It also explores the role of the BM tumor microenvironment as a secure 'niche' for lymphoma cells to grow, proliferate and survive. It further evaluates potential mechanisms through which the tumor cells can establish molecular connections with the BM cells to provide pro-tumor benefits, and discusses putative therapeutic strategies for disrupting the BM-lymphoma cell communication.
Collapse
Affiliation(s)
| | | | | | | | | | - Lalit Sehgal
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; (A.S.); (S.M.C.); (A.H.); (W.C.B.); (S.S.)
| | - Narendranath Epperla
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; (A.S.); (S.M.C.); (A.H.); (W.C.B.); (S.S.)
| |
Collapse
|
12
|
Mathur D, Root AR, Bugaj-Gaweda B, Bisulco S, Tan X, Fang W, Kearney JC, Lucas J, Guffroy M, Golas J, Rohde CM, Stevens C, Kamperschroer C, Kelleher K, Lawrence-Henderson RF, Upeslacis E, Yao J, Narula J, LaVallie ER, Fernandez DR, Buetow BS, Rosfjord E, Bloom L, King LE, Tchistiakova L, Nguyen A, Sapra P. A Novel GUCY2C-CD3 T-Cell Engaging Bispecific Construct (PF-07062119) for the Treatment of Gastrointestinal Cancers. Clin Cancer Res 2020; 26:2188-2202. [DOI: 10.1158/1078-0432.ccr-19-3275] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/19/2019] [Accepted: 01/27/2020] [Indexed: 12/24/2022]
|
13
|
Han S, Huang K, Gu Z, Wu J. Tumor immune microenvironment modulation-based drug delivery strategies for cancer immunotherapy. NANOSCALE 2020; 12:413-436. [PMID: 31829394 DOI: 10.1039/c9nr08086d] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The past years have witnessed promising clinical feedback for anti-cancer immunotherapies, which have become one of the hot research topics; however, they are limited by poor delivery kinetics, narrow patient response profiles, and systemic side effects. To the best of our knowledge, the development of cancer is highly associated with the immune system, especially the tumor immune microenvironment (TIME). Based on the comprehensive understanding of the complexity and diversity of TIME, drug delivery strategies focused on the modulation of TIME can be of great significance for directing and improving cancer immunotherapy. This review highlights the TIME modulation in cancer immunotherapy and summarizes the versatile TIME modulation-based cancer immunotherapeutic strategies, medicative principles and accessory biotechniques for further clinical transformation. Remarkably, the recent advances of cancer immunotherapeutic drug delivery systems and future prospects of TIME modulation-based drug delivery systems for much more controlled and precise cancer immunotherapy will be emphatically discussed.
Collapse
Affiliation(s)
- Shuyan Han
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, PR China.
| | | | | | | |
Collapse
|
14
|
Panowski SH, Kuo TC, Zhang Y, Chen A, Geng T, Aschenbrenner L, Kamperschroer C, Pascua E, Chen W, Delaria K, Farias S, Bateman M, Dushin RG, Chin SM, Van Blarcom TJ, Yeung YA, Lindquist KC, Chunyk AG, Kuang B, Han B, Mirsky M, Pardo I, Buetow B, Martin TG, Wolf JL, Shelton D, Rajpal A, Strop P, Chaparro-Riggers J, Sasu BJ. Preclinical Efficacy and Safety Comparison of CD3 Bispecific and ADC Modalities Targeting BCMA for the Treatment of Multiple Myeloma. Mol Cancer Ther 2019; 18:2008-2020. [DOI: 10.1158/1535-7163.mct-19-0007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 06/03/2019] [Accepted: 08/12/2019] [Indexed: 11/16/2022]
|
15
|
Acheampong DO. Bispecific Antibody (bsAb) Construct Formats and their Application in Cancer Therapy. Protein Pept Lett 2019; 26:479-493. [DOI: 10.2174/0929866526666190311163820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 02/28/2019] [Accepted: 03/03/2019] [Indexed: 12/15/2022]
Abstract
Development of cancers mostly involves more than one signal pathways, because of the complicated nature of cancer cells. As such, the most effective treatment option is the one that stops the cancer cells in their tracks by targeting these signal pathways simultaneously. This explains why therapeutic monoclonal antibodies targeted at cancers exert utmost activity when two or more are used as combination therapy. This notwithstanding, studies elsewhere have proven that when bispecific antibody (bsAb) is engineered from two conventional monoclonal antibodies or their chains, it produces better activity than when used as combination therapy. This therefore presents bispecific antibody (bsAb) as the appropriate and best therapeutic agent for the treatment of such cancers. This review therefore discusses the various engineering formats for bispecific antibodies (bsAbs) and their applications.
Collapse
Affiliation(s)
- Desmond O. Acheampong
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Science, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
16
|
Crisci S, Di Francia R, Mele S, Vitale P, Ronga G, De Filippi R, Berretta M, Rossi P, Pinto A. Overview of Targeted Drugs for Mature B-Cell Non-hodgkin Lymphomas. Front Oncol 2019; 9:443. [PMID: 31214498 PMCID: PMC6558009 DOI: 10.3389/fonc.2019.00443] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 05/09/2019] [Indexed: 12/15/2022] Open
Abstract
The improved knowledge of pathogenetic mechanisms underlying lymphomagenesis and the discovery of the critical role of tumor microenvironments have enabled the design of new drugs against cell targets and pathways. The Food and Drug Administration (FDA) has approved several monoclonal antibodies (mAbs) and small molecule inhibitors (SMIs) for targeted therapy in hematology. This review focuses on the efficacy results of the currently available targeted agents and recaps the main ongoing trials in the setting of mature B-Cell non-Hodgkin lymphomas. The objective is to summarize the different classes of novel agents approved for mature B-cell lymphomas, to describe in synoptic tables the results they achieved and, finally, to draw future scenarios as we glimpse through the ongoing clinical trials. Characteristics and therapeutic efficacy are summarized for the currently approved mAbs [i.e., anti-Cluster of differentiation (CD) mAbs, immune checkpoint inhibitors, chimeric antigen receptor (CAR) T-cell therapy, and bispecific antibodies] as well as for SMIs i.e., inhibitors of B-cell receptor signaling, proteasome, mTOR BCL-2 HDAC pathways. The biological disease profiling of B-cell lymphoma subtypes may foster the discovery of innovative drug strategies for improving survival outcome in lymphoid neoplasms, as well as the trade-offs between efficacy and toxicity. The hope for clinical advantages should carefully be coupled with mindful awareness of the potential pitfalls and the occurrence of uneven, sometimes severe, toxicities.
Collapse
Affiliation(s)
- Stefania Crisci
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori, Fondazione “G. Pascale” IRCCS, Naples, Italy
| | - Raffaele Di Francia
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori, Fondazione “G. Pascale” IRCCS, Naples, Italy
| | - Sara Mele
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori, Fondazione “G. Pascale” IRCCS, Naples, Italy
| | - Pasquale Vitale
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori, Fondazione “G. Pascale” IRCCS, Naples, Italy
| | - Giuseppina Ronga
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori, Fondazione “G. Pascale” IRCCS, Naples, Italy
| | - Rosaria De Filippi
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | | | - Paola Rossi
- Department of Biology and Biotechnology “L. Spallanzani,” University of Pavia, Pavia, Italy
| | - Antonio Pinto
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori, Fondazione “G. Pascale” IRCCS, Naples, Italy
| |
Collapse
|
17
|
Betts A, Haddish-Berhane N, Shah DK, van der Graaf PH, Barletta F, King L, Clark T, Kamperschroer C, Root A, Hooper A, Chen X. A Translational Quantitative Systems Pharmacology Model for CD3 Bispecific Molecules: Application to Quantify T Cell-Mediated Tumor Cell Killing by P-Cadherin LP DART ®. AAPS J 2019; 21:66. [PMID: 31119428 PMCID: PMC6531394 DOI: 10.1208/s12248-019-0332-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/08/2019] [Indexed: 01/12/2023] Open
Abstract
CD3 bispecific antibody constructs recruit cytolytic T cells to kill tumor cells, offering a potent approach to treat cancer. T cell activation is driven by the formation of a trimolecular complex (trimer) between drugs, T cells, and tumor cells, mimicking an immune synapse. A translational quantitative systems pharmacology (QSP) model is proposed for CD3 bispecific molecules capable of predicting trimer concentration and linking it to tumor cell killing. The model was used to quantify the pharmacokinetic (PK)/pharmacodynamic (PD) relationship of a CD3 bispecific targeting P-cadherin (PF-06671008). It describes the disposition of PF-06671008 in the central compartment and tumor in mouse xenograft models, including binding to target and T cells in the tumor to form the trimer. The model incorporates T cell distribution to the tumor, proliferation, and contraction. PK/PD parameters were estimated for PF-06671008 and a tumor stasis concentration (TSC) was calculated as an estimate of minimum efficacious trimer concentration. TSC values ranged from 0.0092 to 0.064 pM across mouse tumor models. The model was translated to the clinic and used to predict the disposition of PF-06671008 in patients, including the impact of binding to soluble P-cadherin. The predicted terminal half-life of PF-06671008 in the clinic was approximately 1 day, and P-cadherin expression and number of T cells in the tumor were shown to be sensitive parameters impacting clinical efficacy. A translational QSP model is presented for CD3 bispecific molecules, which integrates in silico, in vitro and in vivo data in a mechanistic framework, to quantify and predict efficacy across species.
Collapse
Affiliation(s)
- Alison Betts
- Department of Biomedicine Design, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts, 02139, USA.
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, 2300 RA, Leiden, The Netherlands.
| | | | - Dhaval K Shah
- Department of Pharmaceutical Sciences, 455 Kapoor Hall, University at Buffalo, The State University of New York, Buffalo, New York, 14214-8033, USA
| | - Piet H van der Graaf
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, 2300 RA, Leiden, The Netherlands
| | - Frank Barletta
- Oncology Research Unit, Pfizer Inc., 401 N Middletown Rd., Pearl River, New York, 10965, USA
| | - Lindsay King
- Department of Biomedicine Design, Pfizer Inc., 1 Burtt Road, Andover, Massachusetts, USA
| | - Tracey Clark
- Established Med Business, Pfizer Inc., Eastern Point Rd, Groton, Connecticut, 06340, USA
| | - Cris Kamperschroer
- Department of Immunotoxicology, Pfizer Inc., 558 Eastern Point Road, Groton, Connecticut, 06340, USA
| | - Adam Root
- Department of Biomedicine Design, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts, 02139, USA
| | - Andrea Hooper
- Oncology Research Unit, Pfizer Inc., 401 N Middletown Rd., Pearl River, New York, 10965, USA
| | - Xiaoying Chen
- Department of Clinical Pharmacology, Pfizer Inc., 10555 Science Center Dr., San Diego, California, 92121, USA
| |
Collapse
|
18
|
Suurs FV, Lub-de Hooge MN, de Vries EGE, de Groot DJA. A review of bispecific antibodies and antibody constructs in oncology and clinical challenges. Pharmacol Ther 2019; 201:103-119. [PMID: 31028837 DOI: 10.1016/j.pharmthera.2019.04.006] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/27/2019] [Indexed: 01/06/2023]
Abstract
Bispecific antibodies (bsAbs) are antibodies that bind two distinct epitopes to cancer.. For use in oncology, one bsAb has been approved and 57 bsAbs are in clinical trials, none of which has reached phase 3. These bsAbs show great variability in design and mechanism of action. The various designs are often linked to the mechanisms of actions. The majority of bsAbs engage immune cells to destroy tumor cells. However, some bsAbs are also used to deliver payloads to tumors or to block tumor signaling pathways. This review provides insight into the choice of construct for bsAbs, summarizes the clinical development of bsAbs in oncology and identifies subsequent challenges.
Collapse
Affiliation(s)
- Frans V Suurs
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Derk Jan A de Groot
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
19
|
Enhancing bioactivity, physicochemical, and pharmacokinetic properties of a nano-sized, anti-VEGFR2 Adnectin, through PASylation technology. Sci Rep 2019; 9:2978. [PMID: 30814652 PMCID: PMC6393559 DOI: 10.1038/s41598-019-39776-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/01/2019] [Indexed: 02/07/2023] Open
Abstract
The crucial role of VEGF receptor 2 (VEGFR2) signaling in the angiogenesis and metastasis of solid tumors has prompted the development of inhibitors with minimal bystander effects. Recently, Adnectin C has attracted attention for cancer treatment. To overcome the problematic properties of Adnectin, a novel form of Adnectin C has been designed by its fusion to a biodegradable polymeric peptide containing Pro/Ala/Ser (PAS) repetitive residues. E. coli-expressed recombinant fused and unfused proteins were compared in terms of bioactivity, physicochemical, and pharmacokinetic properties using standard methods. Dynamic light scattering (DLS) analysis of PASylated adnectin C revealed an approximate 2-fold increase in particle size with a slight change in the net charge. Additionally, fusion of the PAS sequence improved its stability against the growth of thermo-induced aggregated forms. The high receptor-binding and improved binding kinetic parameters of PASylated Adnectin C was confirmed by ELISA and surface plasmon resonance assays, respectively. Pharmacokinetic studies showed a noticeable increase in the terminal half-life of Adnectin C-PAS#1(200) by a factor of 4.57 after single dose by intravenous injection into female BALB/c mice. The results suggest that PASylation could offer a superior delivery strategy for developing Adnectin-derived drugs with improved patient compliance.
Collapse
|
20
|
Chen S, Li L, Zhang F, Wang Y, Hu Y, Zhao L. Immunoglobulin Gamma-Like Therapeutic Bispecific Antibody Formats for Tumor Therapy. J Immunol Res 2019; 2019:4516041. [PMID: 30886871 PMCID: PMC6388348 DOI: 10.1155/2019/4516041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 11/08/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022] Open
Abstract
Bispecific antibodies (BsAbs) are a sort of dual functional proteins with specific binding to two distinct targets, which have become a focus of interest in antibody engineering and drug development research and have a promising future for wide applications in cancer immunotherapy and autoimmune disease. The key of clinical application and commercial-scale manufacturing of BsAbs is the amenability to assembly and purification of desired heterodimers. Advances in genetic engineering technology had resulted in the development of diverse BsAbs. Multiple recombinant strategies have been used to solve the mispairing problem between light and heavy chains, as well as to enforce accurate dimerization of heterologous heavy chains. There are 23 platforms available to generate 62 BsAbs which can be further divided into IgG-like ones and fragment-based ones, and more than 50 molecules are undergoing clinical trials currently. BsAbs with IgG-like architecture exhibit superior advantages in structure (similar to natural antibodies), pharmacokinetics, half-life, FcR-mediated function, and biological activity. This review considers various IgG-like BsAb generation approaches, summarizes the clinical applications of promising new BsAbs, and describes the mechanism of BsAbs in tumor therapy.
Collapse
Affiliation(s)
- Shixue Chen
- National Clinical Research Center for Normal Aging and Geriatric & Department of Oncology & Institute of Geriatric & The Key Lab of Normal Aging and Geriatric, The Second Medical Centre, PLA General Hospital, Beijing, China
| | - Lingling Li
- National Clinical Research Center for Normal Aging and Geriatric & Department of Oncology & Institute of Geriatric & The Key Lab of Normal Aging and Geriatric, The Second Medical Centre, PLA General Hospital, Beijing, China
- Medical of School & Graduate School, Nankai University, Tianjin, China
| | - Fan Zhang
- National Clinical Research Center for Normal Aging and Geriatric & Department of Oncology & Institute of Geriatric & The Key Lab of Normal Aging and Geriatric, The Second Medical Centre, PLA General Hospital, Beijing, China
| | - Yu Wang
- National Clinical Research Center for Normal Aging and Geriatric & Department of Oncology & Institute of Geriatric & The Key Lab of Normal Aging and Geriatric, The Second Medical Centre, PLA General Hospital, Beijing, China
| | - Yi Hu
- National Clinical Research Center for Normal Aging and Geriatric & Department of Oncology & Institute of Geriatric & The Key Lab of Normal Aging and Geriatric, The Second Medical Centre, PLA General Hospital, Beijing, China
| | - Lei Zhao
- National Clinical Research Center for Normal Aging and Geriatric & Department of Oncology & Institute of Geriatric & The Key Lab of Normal Aging and Geriatric, The Second Medical Centre, PLA General Hospital, Beijing, China
| |
Collapse
|
21
|
Pan H, Liu J, Deng W, Xing J, Li Q, Wang Z. Site-specific PEGylation of an anti-CEA/CD3 bispecific antibody improves its antitumor efficacy. Int J Nanomedicine 2018; 13:3189-3201. [PMID: 29881272 PMCID: PMC5985803 DOI: 10.2147/ijn.s164542] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Introduction Bispecific antibodies that engage immune cells to kill cancer cells are actively pursued in cancer immunotherapy. Different types of bispecific antibodies, including single-chain fragments, Fab fragments, nanobodies, and immunoglobulin Gs (IgGs), have been studied. However, the low molecular weight of bispecific antibodies with single-chain or Fab fragments generally leads to their rapid clearance in vivo, which limits the therapeutic potential of these bispecific antibodies. Materials and methods In this study, we used a site-specific PEGylation strategy to modify the bispecific single-domain antibody-linked Fab (S-Fab), which was designed by linking an anticarcinoembryonic antigen (anti-CEA) nanobody with an anti-CD3 Fab. Results The half-life (t1/2) of PEGylated S-Fab (polyethylene glycol-S-Fab) was increased 12-fold in vivo with a slightly decreased tumor cell cytotoxicity in vitro as well as more potent tumor growth inhibition in vivo compared to S-Fab. Conclusion This study demonstrated that PEGylation is an effective approach to enhance the antitumor efficacy of bispecific antibodies.
Collapse
Affiliation(s)
- Haitao Pan
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Jiayu Liu
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Wentong Deng
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Jieyu Xing
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Qing Li
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zhong Wang
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| |
Collapse
|
22
|
Tong B, Wang M. CD47 is a novel potent immunotherapy target in human malignancies: current studies and future promises. Future Oncol 2018; 14:2179-2188. [PMID: 29667847 DOI: 10.2217/fon-2018-0035] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recently, many immunosuppressive checkpoints such as PD-L1, CTLA-4 and CD47, were identified in succession and serve as potential immunotherapy targets in human cancers. Among them, CD47, a 'marker-of-self' protein that is overexpressed broadly across tumor types, is emerging as a novel potent macrophage immune checkpoint for cancer immunotherapy. In this review, we highlight the prominent role of CD47 as a 'don't-eat-me' signal that inhibits macrophage phagocytosis for immune evasion of a tumor and presents the opportunities and challenges for CD47 inhibitors both as monotherapy and in combination treatments for hematological cancers and solid tumors; some of these agents are currently in clinical trials.
Collapse
Affiliation(s)
- Bing Tong
- Lung Cancer Center, Department of Respiratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China; Postal address: No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, PR China
| | - Mengzhao Wang
- Lung Cancer Center, Department of Respiratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China; Postal address: No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, PR China
| |
Collapse
|
23
|
Manoutcharian K, Perez-Garmendia R, Gevorkian G. Recombinant Antibody Fragments for Neurodegenerative Diseases. Curr Neuropharmacol 2018; 15:779-788. [PMID: 27697033 PMCID: PMC5771054 DOI: 10.2174/1570159x01666160930121647] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/04/2016] [Accepted: 09/28/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Recombinant antibody fragments are promising alternatives to full-length immunoglobulins and offer important advantages compared with conventional monoclonal antibodies: extreme specificity, higher affinity, superior stability and solubility, reduced immunogenicity as well as easy and inexpensive large-scale production. OBJECTIVE In this article we will review and discuss recombinant antibodies that are being evaluated for neurodegenerative diseases in pre-clinical models and in clinical studies and will summarize new strategies that are being developed to optimize their stability, specificity and potency for advancing their use. METHODS Articles describing recombinant antibody fragments used for neurological diseases were selected (PubMed) and evaluated for their significance. RESULTS Different antibody formats such as single-chain fragment variable (scFv), single-domain antibody fragments (VHHs or sdAbs), bispecific antibodies (bsAbs), intrabodies and nanobodies, are currently being studied in pre-clinical models of cancer as well as infectious and autoimmune diseases and many of them are being tested as therapeutics in clinical trials. Immunotherapy approaches have shown therapeutic efficacy in several animal models of Alzheimer´s disease (AD), Parkinson disease (PD), dementia with Lewy bodies (DLB), frontotemporal dementia (FTD), Huntington disease (HD), transmissible spongiform encephalopathies (TSEs) and multiple sclerosis (MS). It has been demonstrated that recombinant antibody fragments may neutralize toxic extra- and intracellular misfolded proteins involved in the pathogenesis of AD, PD, DLB, FTD, HD or TSEs and may target toxic immune cells participating in the pathogenesis of MS. CONCLUSION Recombinant antibody fragments represent a promising tool for the development of antibody-based immunotherapeutics for neurodegenerative diseases.
Collapse
Affiliation(s)
- Karen Manoutcharian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Mexico DF. Mexico
| | - Roxanna Perez-Garmendia
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Mexico DF. Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Apartado Postal 70228, Cuidad Universitaria, Mexico DF, CP 04510, Mexico. 0
| |
Collapse
|
24
|
Abstract
The question of whether human tumors express antigens that can be recognized by the immune system has been answered with a resounding YES. Most were identified through spontaneous antitumor humoral and cellular immune responses found in cancer patients and include peptides, glycopeptides, phosphopeptides, viral peptides, and peptides resulting from common mutations in oncogenes and tumor-suppressor genes, or common gene fusion events. Many have been extensively tested as candidates for anticancer vaccines. More recently, attention has been focused on the potentially large number of unique tumor antigens, mutated neoantigens, that are the predicted products of the numerous mutations revealed by exome sequencing of primary tumors. Only a few have been confirmed as targets of spontaneous immunity and immunosurveillance, and even fewer have been tested in preclinical and clinical settings. The field has been divided for a long time on the relative importance of shared versus mutated antigens in tumor surveillance and as candidates for vaccines. This question will eventually need to be answered in a head to head comparison in well-designed clinical trials. One advantage that shared antigens have over mutated antigens is their potential to be used in vaccines for primary cancer prevention. Cancer Immunol Res; 5(5); 347-54. ©2017 AACR.
Collapse
Affiliation(s)
- Olivera J Finn
- Department of Immunology, University of Pittsburgh School of Medicine and the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.
| |
Collapse
|
25
|
Fisher TS, Hooper AT, Lucas J, Clark TH, Rohner AK, Peano B, Elliott MW, Tsaparikos K, Wang H, Golas J, Gavriil M, Haddish-Berhane N, Tchistiakova L, Gerber HP, Root AR, May C. A CD3-bispecific molecule targeting P-cadherin demonstrates T cell-mediated regression of established solid tumors in mice. Cancer Immunol Immunother 2018; 67:247-259. [PMID: 29067496 PMCID: PMC11028296 DOI: 10.1007/s00262-017-2081-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/14/2017] [Indexed: 12/11/2022]
Abstract
Strong evidence exists supporting the important role T cells play in the immune response against tumors. Still, the ability to initiate tumor-specific immune responses remains a challenge. Recent clinical trials suggest that bispecific antibody-mediated retargeted T cells are a promising therapeutic approach to eliminate hematopoietic tumors. However, this approach has not been validated in solid tumors. PF-06671008 is a dual-affinity retargeting (DART®)-bispecific protein engineered with enhanced pharmacokinetic properties to extend in vivo half-life, and designed to engage and activate endogenous polyclonal T cell populations via the CD3 complex in the presence of solid tumors expressing P-cadherin. This bispecific molecule elicited potent P-cadherin expression-dependent cytotoxic T cell activity across a range of tumor indications in vitro, and in vivo in tumor-bearing mice. Regression of established tumors in vivo was observed in both cell line and patient-derived xenograft models engrafted with circulating human T lymphocytes. Measurement of in vivo pharmacodynamic markers demonstrates PF-06671008-mediated T cell activation, infiltration and killing as the mechanism of tumor inhibition.
Collapse
Affiliation(s)
- Timothy S Fisher
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA.
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA.
- Pfizer Inc., 10777 Science Center Drive, San Diego, CA, 92121, USA.
| | - Andrea T Hooper
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Justin Lucas
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | | | - Allison K Rohner
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Bryan Peano
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Mark W Elliott
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Konstantinos Tsaparikos
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Hui Wang
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Jonathan Golas
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Maria Gavriil
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Nahor Haddish-Berhane
- BioMedicine Design Pfizer Inc., Cambridge, MA, USA
- Johnson and Johnson Pharmaceutical Research and Development, Spring House, PA, USA
| | | | - Hans-Peter Gerber
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
- Maverick Therapeutics, Brisbane, CA, USA
| | - Adam R Root
- BioMedicine Design Pfizer Inc., Cambridge, MA, USA
| | - Chad May
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
- Maverick Therapeutics, Brisbane, CA, USA
| |
Collapse
|
26
|
Current Status of Immuno-Oncology in Hematologic Cancers. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
27
|
Brinkmann U, Kontermann RE. The making of bispecific antibodies. MAbs 2017; 9:182-212. [PMID: 28071970 PMCID: PMC5297537 DOI: 10.1080/19420862.2016.1268307] [Citation(s) in RCA: 647] [Impact Index Per Article: 80.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022] Open
Abstract
During the past two decades we have seen a phenomenal evolution of bispecific antibodies for therapeutic applications. The 'zoo' of bispecific antibodies is populated by many different species, comprising around 100 different formats, including small molecules composed solely of the antigen-binding sites of two antibodies, molecules with an IgG structure, and large complex molecules composed of different antigen-binding moieties often combined with dimerization modules. The application of sophisticated molecular design and genetic engineering has solved many of the technical problems associated with the formation of bispecific antibodies such as stability, solubility and other parameters that confer drug properties. These parameters may be summarized under the term 'developability'. In addition, different 'target product profiles', i.e., desired features of the bispecific antibody to be generated, mandates the need for access to a diverse panel of formats. These may vary in size, arrangement, valencies, flexibility and geometry of their binding modules, as well as in their distribution and pharmacokinetic properties. There is not 'one best format' for generating bispecific antibodies, and no single format is suitable for all, or even most of, the desired applications. Instead, the bispecific formats collectively serve as a valuable source of diversity that can be applied to the development of therapeutics for various indications. Here, a comprehensive overview of the different bispecific antibody formats is provided.
Collapse
Affiliation(s)
- Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Im Nonnenwald, Penzberg, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Nobelstraße, Stuttgart, Germany
| |
Collapse
|
28
|
Abstract
Immunotherapy has shown promise in many solid tumors including melanoma and non-small cell lung cancer with an evolving role in breast cancer. Immunotherapy encompasses a wide range of therapies including immune checkpoint inhibition, monoclonal antibodies, bispecific antibodies, vaccinations, antibody-drug conjugates, and identifying other emerging interventions targeting the tumor microenvironment. Increasing efficacy of these treatments in breast cancer patients requires identification of better biomarkers to guide patient selection; recognizing when to initiate these therapies in multi-modality treatment plans; establishing novel assays to monitor immune-mediated responses; and creating combined systemic therapy options incorporating conventional treatments such as chemotherapy and endocrine therapy. This review will focus on the current role and future directions of many of these immunotherapies in breast cancer, as well as highlighting clinical trials that are investigating several of these active issues.
Collapse
|
29
|
de Bruin RCG, Veluchamy JP, Lougheed SM, Schneiders FL, Lopez-Lastra S, Lameris R, Stam AG, Sebestyen Z, Kuball J, Molthoff CFM, Hooijberg E, Roovers RC, Santo JPD, van Bergen En Henegouwen PMP, Verheul HMW, de Gruijl TD, van der Vliet HJ. A bispecific nanobody approach to leverage the potent and widely applicable tumor cytolytic capacity of Vγ9Vδ2-T cells. Oncoimmunology 2017; 7:e1375641. [PMID: 29296532 DOI: 10.1080/2162402x.2017.1375641] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/11/2017] [Accepted: 08/31/2017] [Indexed: 12/23/2022] Open
Abstract
Though Vγ9Vδ2-T cells constitute only a small fraction of the total T cell population in human peripheral blood, they play a vital role in tumor defense and are therefore of major interest to explore for cancer immunotherapy. Vγ9Vδ2-T cell-based cancer immunotherapeutic approaches developed so far have been generally well tolerated and were able to induce significant clinical responses. However, overall results were inconsistent, possibly due to the fact that these strategies induced systemic activation of Vγ9Vδ2-T cells without preferential accumulation and targeted activation in the tumor. Here we show that a novel bispecific nanobody-based construct targeting both Vγ9Vδ2-T cells and EGFR induced potent Vγ9Vδ2-T cell activation and subsequent tumor cell lysis both in vitro and in an in vivo mouse xenograft model. Tumor cell lysis was independent of KRAS and BRAF tumor mutation status and common Vγ9Vδ2-T cell receptor sequence variations. In combination with the conserved monomorphic nature of the Vγ9Vδ2-TCR and the facile replacement of the tumor-specific nanobody, this immunotherapeutic approach can be applied to a large group of cancer patients.
Collapse
Affiliation(s)
- Renée C G de Bruin
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - John P Veluchamy
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Sinéad M Lougheed
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Famke L Schneiders
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Silvia Lopez-Lastra
- Innate Immunity Unit, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1223, Paris, France.,Université Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Roeland Lameris
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Anita G Stam
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Zsolt Sebestyen
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Jürgen Kuball
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Carla F M Molthoff
- Department of Radiology and Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Erik Hooijberg
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Rob C Roovers
- Department of Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1223, Paris, France
| | | | - Henk M W Verheul
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Xenaki KT, Oliveira S, van Bergen En Henegouwen PMP. Antibody or Antibody Fragments: Implications for Molecular Imaging and Targeted Therapy of Solid Tumors. Front Immunol 2017; 8:1287. [PMID: 29075266 PMCID: PMC5643388 DOI: 10.3389/fimmu.2017.01287] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/25/2017] [Indexed: 01/10/2023] Open
Abstract
The use of antibody-based therapeutics has proven very promising for clinical applications in cancer patients, with multiple examples of antibodies and antibody–drug conjugates successfully applied for the treatment of solid tumors and lymphomas. Given reported recurrence rates, improvements are clearly still necessary. A major factor limiting the efficacy of antibody-targeted cancer therapies may be the incomplete penetration of the antibody or antibody–drug conjugate into the tumor. Incomplete tumor penetration also affects the outcome of molecular imaging, when using such targeting agents. From the injection site until they arrive inside the tumor, targeting molecules are faced with several barriers that impact intratumoral distribution. The primary means of antibody transport inside tumors is based on diffusion. The diffusive penetration inside the tumor is influenced by both antibody properties, such as size and binding affinity, as well as tumor properties, such as microenvironment, vascularization, and targeted antigen availability. Engineering smaller antibody fragments has shown to improve the rate of tumor uptake and intratumoral distribution. However, it is often accompanied by more rapid clearance from the body and in several cases also by inherent destabilization and reduction of the binding affinity of the antibody. In this perspective, we discuss different cancer targeting approaches based on antibodies or their fragments. We carefully consider how their size and binding properties influence their intratumoral uptake and distribution, and how this may affect cancer imaging and therapy of solid tumors.
Collapse
Affiliation(s)
- Katerina T Xenaki
- Division of Cell Biology, Science Faculty, Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Sabrina Oliveira
- Division of Cell Biology, Science Faculty, Department of Biology, Utrecht University, Utrecht, Netherlands.,Pharmaceutics, Department of Pharmaceutical Sciences, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | | |
Collapse
|
31
|
Chang CH, Goldenberg DM. Enhancing the antitumor potency of T cells redirected by bispecific antibodies. Oncoscience 2017; 4:120-121. [PMID: 29142899 PMCID: PMC5672892 DOI: 10.18632/oncoscience.366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 01/13/2023] Open
Affiliation(s)
- Chien-Hsing Chang
- IBC Pharmaceuticals Inc., and Immunomedics Inc., Morris Plains, New Jersey, USA
| | - David M Goldenberg
- IBC Pharmaceuticals Inc., and Immunomedics Inc., Morris Plains, New Jersey, USA
| |
Collapse
|
32
|
Zhu M, Wu B, Brandl C, Johnson J, Wolf A, Chow A, Doshi S. Blinatumomab, a Bispecific T-cell Engager (BiTE(®)) for CD-19 Targeted Cancer Immunotherapy: Clinical Pharmacology and Its Implications. Clin Pharmacokinet 2017; 55:1271-1288. [PMID: 27209293 DOI: 10.1007/s40262-016-0405-4] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Blinatumomab is a bispecific T-cell engager (BiTE(®)) antibody construct that transiently links CD19-positive B cells to CD3-positive T cells, resulting in induction of T-cell-mediated serial lysis of B cells and concomitant T-cell proliferation. Blinatumomab showed anti-leukemia activity in clinical trials and was approved by the US Food and Drug Administration for the treatment of Philadelphia chromosome-negative relapsed/refractory B-cell precursor acute lymphoblastic leukemia (r/r ALL). The objectives of this work were to characterize blinatumomab pharmacokinetics and pharmacodynamics and to evaluate dosing regimens. METHODS Data from six phase I and II trials in patients with r/r ALL, minimal residual disease-positive ALL, and non-Hodgkin's lymphoma (NHL) were analyzed. Blinatumomab pharmacokinetics was characterized by non-compartmental and population pharmacokinetic analyses and pharmacodynamics was described graphically. RESULTS Blinatumomab exhibited linear pharmacokinetics under continuous intravenous infusion for 4-8 weeks per cycle over a dose range of 5-90 µg/m(2)/day, without target-mediated disposition. Estimated mean (standard deviation) volume of distribution, clearance, and elimination half-life were 4.52 (2.89) L, 2.72 (2.71) L/h, and 2.11 (1.42) h, respectively. Pharmacokinetics was similar in patients with ALL and NHL and was not affected by patient demographics, supporting fixed dosing in adults. Although creatinine clearance was a significant covariate of drug clearance, no dose adjustment was required in patients with mild or moderate renal impairment. Incidence of neutralizing antidrug antibodies was <1 %. Blinatumomab pharmacodynamics featured T-cell redistribution and activation, B-cell depletion, and transient dose-dependent cytokine elevation. Blinatumomab did not affect cytochrome P450 enzymes directly; cytokines may trigger transient cytochrome P450 suppression with low potential for inducing drug interactions. CONCLUSIONS Blinatumomab has unique pharmacokinetic and immunological features that require indication-dependent dosing regimens. Stepped dosing is required to achieve adequate efficacy and minimize cytokine release in diseases with high tumor burden.
Collapse
Affiliation(s)
- Min Zhu
- Amgen Inc., Thousand Oaks, CA, 91320, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Matsuki E, Younes A. Checkpoint Inhibitors and Other Immune Therapies for Hodgkin and Non-Hodgkin Lymphoma. Curr Treat Options Oncol 2017; 17:31. [PMID: 27193488 DOI: 10.1007/s11864-016-0401-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OPINION STATEMENT Treatment for relapsed/refractory (R/R) Hodgkin and non-Hodgkin lymphoma remains challenging. The introduction of rituximab to B cell non-Hodgkin lymphoma (B-NHL) treatment significantly improved patients' response rate and survival; however, approximately one third of patients with diffuse large B cell lymphoma, the most common B-NHL subtype, still have a relapse or become refractory after first-line therapy. More recently, antibody therapies and small-molecule inhibitors were approved for treating R/R lymphomas; these agents include brentuximab vedotin, ibrutinib, and idelalisib. Immune checkpoint inhibitors and other immune therapies are emerging treatments currently being evaluated in various clinical trials for their efficacy against lymphoid malignancies. Striking results from these treatment modalities have been observed in solid tumors, and evidence is accumulating to support their use in various lymphomas. The most exciting results from immune checkpoint inhibitor therapy have been seen in patients with R/R Hodgkin lymphoma, in whom the overall response rate has reached 60-80 %. Results in NHL are more similar to those seen in other solid malignancies, ranging between 20 and 40 %, depending on the histology. Formal approval of these drugs is being awaited, as are the results of combination therapy with checkpoint inhibitors and other treatment modalities, including conventional chemotherapy, small-molecule inhibitors, and other immune therapies. Although response rates have been promising, attention must be paid to the management of unique immune-related adverse events, which warrant close monitoring in some cases. Identification of biomarkers that predict response or severe adverse events using either the tumor specimen or peripheral blood would aid in selecting patients suited for these types of treatment as well as determining the ideal sequence of treatment within the realm of immune therapies.
Collapse
Affiliation(s)
- Eri Matsuki
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 330, New York, NY, 10065, USA
| | - Anas Younes
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 330, New York, NY, 10065, USA.
| |
Collapse
|
34
|
Xing J, Lin L, Li J, Liu J, Zhou C, Pan H, Shu R, Dong B, Cao D, Li Q, Wang Z. BiHC, a T-Cell-Engaging Bispecific Recombinant Antibody, Has Potent Cytotoxic Activity Against Her2 Tumor Cells. Transl Oncol 2017; 10:780-785. [PMID: 28797938 PMCID: PMC5548338 DOI: 10.1016/j.tranon.2017.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/09/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
Among different cancer immunotherapy approaches, bispecific antibodies (BsAbs) are of great interest due to their ability to recruit immune cells to kill tumor cells directly. Various BsAbs against Her2 tumor cells have been proposed with potent cytotoxic activities. However, most of these formats require extensive processing to obtain heterodimeric bispecific antibodies. In this study, we describe a bispecific antibody, BiHC (bispecific Her2-CD3 antibody), constructed with a single-domain anti-Her2 and a single-chain Fv (variable fragment) of anti-CD3 in an IgG-like format. In contrast to most IgG-like BsAbs, the two arms in BiHC have different molecular weights, making it easier to separate hetero- or homodimers. BiHC can be expressed in Escherichia coli and purified via Protein A affinity chromatography. The purified BiHC can recruit T cells and induce specific cytotoxicity of Her2-expressing tumor cells in vitro. The BiHC can also efficiently inhibit the tumor growth in vivo. Thus, BiHC is a promising candidate for the treatment of Her2-positive cancers.
Collapse
Affiliation(s)
- Jieyu Xing
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Limin Lin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Jing Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Jiayu Liu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Changhua Zhou
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Haitao Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Rui Shu
- Ying Rui Inc., Guangzhou, Guangdong, China 510009.
| | - Bin Dong
- School of Biosciences and Biopharmaceutics, Guangdong pharmaceutical University, Guangzhou, Guangdong, China 510009.
| | - Donglin Cao
- Department of Laboratory Medicine, Guangdong No. 2 Provincial People's Hospital, Guangzhou, China 510317.
| | - Qing Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Zhong Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| |
Collapse
|
35
|
Lameris R, de Bruin RCG, van Bergen En Henegouwen PMP, Verheul HM, Zweegman S, de Gruijl TD, van der Vliet HJ. Generation and characterization of CD1d-specific single-domain antibodies with distinct functional features. Immunology 2017; 149:111-21. [PMID: 27312006 DOI: 10.1111/imm.12635] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/25/2016] [Accepted: 06/12/2016] [Indexed: 12/17/2022] Open
Abstract
Ligation of the CD1d antigen-presenting molecule by monoclonal antibodies (mAbs) can trigger important biological functions. For therapeutic purposes camelid-derived variable domain of heavy-chain-only antibodies (VHH) have multiple advantages over mAbs because they are small, stable and have low immunogenicity. Here, we generated 21 human CD1d-specific VHH by immunizing Lama glama and subsequent phage display. Two clones induced maturation of dendritic cells, one clone induced early apoptosis in CD1d-expressing B lymphoblasts and multiple myeloma cells, and another clone blocked recognition of glycolipid-loaded CD1d by CD1d-restricted invariant natural killer T (iNKT) cells. In contrast to reported CD1d-specific mAbs, these CD1d-specific VHH have the unique characteristic that they induce specific and well-defined biological effects. This feature, combined with the above-indicated general advantages of VHH, make the CD1d-specific VHH generated here unique and useful tools to exploit both CD1d ligation as well as disruption of CD1d-iNKT interactions in the treatment of cancer or inflammatory disorders.
Collapse
Affiliation(s)
- Roeland Lameris
- Department of Medical Oncology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Renée C G de Bruin
- Department of Medical Oncology, VU University Medical Centre, Amsterdam, the Netherlands
| | | | - Henk M Verheul
- Department of Medical Oncology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Sonja Zweegman
- Department of Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology, VU University Medical Centre, Amsterdam, the Netherlands
| |
Collapse
|
36
|
Trivedi A, Stienen S, Zhu M, Li H, Yuraszeck T, Gibbs J, Heath T, Loberg R, Kasichayanula S. Clinical Pharmacology and Translational Aspects of Bispecific Antibodies. Clin Transl Sci 2017; 10:147-162. [PMID: 28297195 PMCID: PMC5421745 DOI: 10.1111/cts.12459] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/30/2017] [Indexed: 02/07/2023] Open
Affiliation(s)
- A Trivedi
- Amgen Inc., Thousand Oaks, California, USA
| | - S Stienen
- Amgen Research (Munich), Munich, Germany
| | - M Zhu
- Amgen Inc., Thousand Oaks, California, USA
| | - H Li
- Amgen Inc., Thousand Oaks, California, USA
| | | | - J Gibbs
- Amgen Inc., Thousand Oaks, California, USA.,Current address: AbbVie Inc., North Chicago, Illinois, USA
| | - T Heath
- Amgen Inc., Thousand Oaks, California, USA
| | - R Loberg
- Amgen Inc., Thousand Oaks, California, USA
| | - S Kasichayanula
- Amgen Inc., Thousand Oaks, California, USA.,Current Address: AbbVie Inc., South San Francisco, California, USA
| |
Collapse
|
37
|
Abstract
In this issue of Cancer Cell, Li et al. and Seckinger et al. describe promising results of two T-cell-dependent bi-specific antibodies for the treatment of multiple myeloma: one targets FcRH5 expressed on B cells, whereas the other targets the B cell maturation antigen expressed on plasma cells.
Collapse
Affiliation(s)
- Marta Chesi
- Mayo Clinic in Arizona, Phoenix, AZ 85054, USA
| | | |
Collapse
|
38
|
Seckinger A, Delgado JA, Moser S, Moreno L, Neuber B, Grab A, Lipp S, Merino J, Prosper F, Emde M, Delon C, Latzko M, Gianotti R, Lüoend R, Murr R, Hosse RJ, Harnisch LJ, Bacac M, Fauti T, Klein C, Zabaleta A, Hillengass J, Cavalcanti-Adam EA, Ho AD, Hundemer M, San Miguel JF, Strein K, Umaña P, Hose D, Paiva B, Vu MD. Target Expression, Generation, Preclinical Activity, and Pharmacokinetics of the BCMA-T Cell Bispecific Antibody EM801 for Multiple Myeloma Treatment. Cancer Cell 2017; 31:396-410. [PMID: 28262554 DOI: 10.1016/j.ccell.2017.02.002] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 10/07/2016] [Accepted: 01/31/2017] [Indexed: 01/12/2023]
Abstract
We identified B cell maturation antigen (BCMA) as a potential therapeutic target in 778 newly diagnosed and relapsed myeloma patients. We constructed an IgG-based BCMA-T cell bispecific antibody (EM801) and showed that it increased CD3+ T cell/myeloma cell crosslinking, followed by CD4+/CD8+ T cell activation, and secretion of interferon-γ, granzyme B, and perforin. This effect is CD4 and CD8 T cell mediated. EM801 induced, at nanomolar concentrations, myeloma cell death by autologous T cells in 34 of 43 bone marrow aspirates, including those from high-risk patients and patients after multiple lines of treatment, tumor regression in six of nine mice in a myeloma xenograft model, and depletion of BCMA+ cells in cynomolgus monkeys. Pharmacokinetics and pharmacodynamics indicate weekly intravenous/subcutaneous administration.
Collapse
Affiliation(s)
- Anja Seckinger
- Universitätsklinikum Heidelberg, Labor für Myelomforschung and Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Jose Antonio Delgado
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | - Samuel Moser
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Laura Moreno
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | - Brigitte Neuber
- Universitätsklinikum Heidelberg, Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Anna Grab
- Universitätsklinikum Heidelberg, Labor für Myelomforschung and Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Susanne Lipp
- Universitätsklinikum Heidelberg, Labor für Myelomforschung and Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Juana Merino
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | - Felipe Prosper
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | - Martina Emde
- Universitätsklinikum Heidelberg, Labor für Myelomforschung and Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Camille Delon
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Melanie Latzko
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Reto Gianotti
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Remo Lüoend
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Ramona Murr
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Ralf J Hosse
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | | | - Marina Bacac
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Tanja Fauti
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | | | - Aintzane Zabaleta
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | - Jens Hillengass
- Universitätsklinikum Heidelberg, Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Elisabetta Ada Cavalcanti-Adam
- University of Heidelberg, Institute for Physical Chemistry and Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Anthony D Ho
- Universitätsklinikum Heidelberg, Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Michael Hundemer
- Universitätsklinikum Heidelberg, Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Jesus F San Miguel
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | | | - Pablo Umaña
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Dirk Hose
- Universitätsklinikum Heidelberg, Labor für Myelomforschung and Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | - Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain.
| | | |
Collapse
|
39
|
Eckert F, Gaipl U, Niedermann G, Hettich M, Schilbach K, Huber S, Zips D. Beyond checkpoint inhibition - Immunotherapeutical strategies in combination with radiation. Clin Transl Radiat Oncol 2017; 2:29-35. [PMID: 29657997 PMCID: PMC5893529 DOI: 10.1016/j.ctro.2016.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 12/20/2022] Open
Abstract
The revival of cancer immunotherapy has taken place with the clinical success of immune checkpoint inhibition. However, the spectrum of immunotherapeutic approaches is much broader encompassing T cell engaging strategies, tumour-specific vaccination, antibodies or immunocytokines. This review focuses on the immunological effects of irradiation and the evidence available on combination strategies with immunotherapy. The available data suggest great potential of combined treatments, yet also poses questions about dose, fractionation, timing and most promising multimodal strategies.
Collapse
Key Words
- Bispecific antibodies
- CAR, chimeric antigen receptor
- CAR-T-cells
- CDN, cyclic dinucleotides
- CTL, cytotoxic T lymphocyte
- CTLA-4, cytotoxic T-lymphocyte-associated protein 4
- GM-CSF, granulocyte-monocyte colony stimulating factor
- IR, irradiation
- Immunocytokines
- Immunotherapy
- PD-1, Programmed cell death protein 1 receptor
- PD-L1, PD-1 ligand
- Radiotherapy
- TCR, T cell receptor
- Treg, regulatory T cells
- Vaccination
- bsAb, bispecific antibody
- scFv, single chain variable fragment
Collapse
Affiliation(s)
- F. Eckert
- Department of Radiation Oncology, Universitaetsklinikum Tuebingen, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | - U.S. Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - G. Niedermann
- Department of Radiation Oncology, Medical Center – University of Freiburg, Freiburg, Germany
| | - M. Hettich
- Department of Radiation Oncology, Medical Center – University of Freiburg, Freiburg, Germany
| | - K. Schilbach
- Department of General Pediatrics/Pediatric Oncology, Universitaetsklinikum Tuebingen, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | - S.M. Huber
- Department of Radiation Oncology, Universitaetsklinikum Tuebingen, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | - D. Zips
- Department of Radiation Oncology, Universitaetsklinikum Tuebingen, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| |
Collapse
|
40
|
de Bruin RCG, Stam AGM, Vangone A, van Bergen En Henegouwen PMP, Verheul HMW, Sebestyén Z, Kuball J, Bonvin AMJJ, de Gruijl TD, van der Vliet HJ. Prevention of Vγ9Vδ2 T Cell Activation by a Vγ9Vδ2 TCR Nanobody. THE JOURNAL OF IMMUNOLOGY 2016; 198:308-317. [PMID: 27895170 DOI: 10.4049/jimmunol.1600948] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 11/02/2016] [Indexed: 01/09/2023]
Abstract
Vγ9Vδ2 T cell activation plays an important role in antitumor and antimicrobial immune responses. However, there are conditions in which Vγ9Vδ2 T cell activation can be considered inappropriate for the host. Patients treated with aminobisphosphonates for hypercalcemia or metastatic bone disease often present with a debilitating acute phase response as a result of Vγ9Vδ2 T cell activation. To date, no agents are available that can clinically inhibit Vγ9Vδ2 T cell activation. In this study, we describe the identification of a single domain Ab fragment directed to the TCR of Vγ9Vδ2 T cells with neutralizing properties. This variable domain of an H chain-only Ab (VHH or nanobody) significantly inhibited both phosphoantigen-dependent and -independent activation of Vγ9Vδ2 T cells and, importantly, strongly reduced the production of inflammatory cytokines upon stimulation with aminobisphosphonate-treated cells. Additionally, in silico modeling suggests that the neutralizing VHH binds the same residues on the Vγ9Vδ2 TCR as the Vγ9Vδ2 T cell Ag-presenting transmembrane protein butyrophilin 3A1, providing information on critical residues involved in this interaction. The neutralizing Vγ9Vδ2 TCR VHH identified in this study might provide a novel approach to inhibit the unintentional Vγ9Vδ2 T cell activation as a consequence of aminobisphosphonate administration.
Collapse
Affiliation(s)
- Renée C G de Bruin
- Department of Medical Oncology, VU University Medical Center, 1081 HV Amsterdam, the Netherlands
| | - Anita G M Stam
- Department of Medical Oncology, VU University Medical Center, 1081 HV Amsterdam, the Netherlands
| | - Anna Vangone
- Bijvoet Center for Biomolecular Research, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | | | - Henk M W Verheul
- Department of Medical Oncology, VU University Medical Center, 1081 HV Amsterdam, the Netherlands
| | - Zsolt Sebestyén
- Laboratory of Translational Immunology, Department of Hematology, University Medical Center Utrecht, 3508 GA Utrecht, the Netherlands
| | - Jürgen Kuball
- Laboratory of Translational Immunology, Department of Hematology, University Medical Center Utrecht, 3508 GA Utrecht, the Netherlands
| | - Alexandre M J J Bonvin
- Bijvoet Center for Biomolecular Research, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center, 1081 HV Amsterdam, the Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology, VU University Medical Center, 1081 HV Amsterdam, the Netherlands;
| |
Collapse
|
41
|
Emerging concepts of T cell metabolism as a target of immunotherapy. Nat Immunol 2016; 17:364-8. [PMID: 27002844 DOI: 10.1038/ni.3415] [Citation(s) in RCA: 258] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 02/17/2016] [Indexed: 12/27/2022]
Abstract
T cells have a pivotal protective role in defense against infection and cancer but also are instrumental in the development of many autoimmune diseases. The regulation of nutrient uptake and utilization in T cells is critically important for the control of their differentiation, and manipulating metabolic pathways in these cells can alter their function and longevity. While the importance of T cell metabolic remodeling in different physiological settings is not fully understood, there is a growing realization that inappropriate metabolic remodeling underlies many aberrant immune responses and that manipulating cellular metabolism can beneficially enhance or temper immunity. Here we comment on the basic metabolic pathways in T cells, followed by a discussion on up-to-date findings about the relationship between metabolism and T cell function and longevity. Furthermore, we expand on potential approaches and applications in which T cells might be manipulated by the reprogramming of metabolic pathways for therapeutic purposes.
Collapse
|
42
|
Chen X, Haddish-Berhane N, Moore P, Clark T, Yang Y, Li H, Xuan D, Barton HA, Betts AM, Barletta F. Mechanistic Projection of First-in-Human Dose for Bispecific Immunomodulatory P-Cadherin LP-DART: An Integrated PK/PD Modeling Approach. Clin Pharmacol Ther 2016; 100:232-41. [DOI: 10.1002/cpt.393] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/05/2016] [Accepted: 05/06/2016] [Indexed: 12/19/2022]
Affiliation(s)
- X Chen
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Cambridge Massachusetts USA
| | - N Haddish-Berhane
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Groton Connecticut USA
- Current Address: Clinical Pharmacology and Pharmacometrics; Quantitative Sciences Janssen Pharmaceuticals, Spring House, Pennsylvania USA
| | - P Moore
- MacroGenics; Rockville; Maryland USA
| | - T Clark
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Groton Connecticut USA
| | - Y Yang
- MacroGenics; Rockville; Maryland USA
| | - H Li
- MacroGenics; Rockville; Maryland USA
| | - D Xuan
- Clinical Pharmacology; Pfizer; San Diego California USA
| | - HA Barton
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Groton Connecticut USA
| | - AM Betts
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Groton Connecticut USA
| | - F Barletta
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Pearl River New York USA
| |
Collapse
|
43
|
de Bruin RCG, Lougheed SM, van der Kruk L, Stam AG, Hooijberg E, Roovers RC, van Bergen En Henegouwen PMP, Verheul HMW, de Gruijl TD, van der Vliet HJ. Highly specific and potently activating Vγ9Vδ2-T cell specific nanobodies for diagnostic and therapeutic applications. Clin Immunol 2016; 169:128-138. [PMID: 27373969 DOI: 10.1016/j.clim.2016.06.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 06/15/2016] [Accepted: 06/29/2016] [Indexed: 01/23/2023]
Abstract
Vγ9Vδ2-T cells constitute the predominant subset of γδ-T cells in human peripheral blood and have been shown to play an important role in antimicrobial and antitumor immune responses. Several efforts have been initiated to exploit these cells for cancer immunotherapy, e.g. by using phosphoantigens, adoptive cell transfer, and by a bispecific monoclonal antibody based approach. Here, we report the generation of a novel set of Vγ9Vδ2-T cell specific VHH (or nanobody). VHH have several advantages compared to conventional antibodies related to their small size, stability, ease of generating multispecific molecules and low immunogenicity. With high specificity and affinity, the anti-Vγ9Vδ2-T cell receptor VHHs are shown to be useful for FACS, MACS and immunocytochemistry. In addition, some VHH were found to specifically activate Vγ9Vδ2-T cells. Besides being of possible immunotherapeutic value, these single domain antibodies will be of great value in the further study of this important immune effector cell subset.
Collapse
Affiliation(s)
- Renée C G de Bruin
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
| | - Sinéad M Lougheed
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
| | - Liza van der Kruk
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
| | - Anita G Stam
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
| | - Erik Hooijberg
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
| | - Rob C Roovers
- Department of Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | | | - Henk M W Verheul
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
| | - Hans J van der Vliet
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
| |
Collapse
|
44
|
Smits NC, Coupet TA, Godbersen C, Sentman CL. Designing multivalent proteins based on natural killer cell receptors and their ligands as immunotherapy for cancer. Expert Opin Biol Ther 2016; 16:1105-12. [PMID: 27248342 DOI: 10.1080/14712598.2016.1195364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Natural killer (NK) cells are an important component of the innate immune system that play a key role in host immunity against cancer. NK cell recognition and activation is based on cell surface receptors recognizing specific ligands that are expressed on many types of tumor cells. Some of these receptors are capable of activating NK cell function while other receptors inhibit NK cell function. Therapeutic approaches to treat cancer have been developed based on preventing NK cell inhibition or using NK cell receptors and their ligands to activate NK cells or T cells to destroy tumor cells. AREAS COVERED This article describes the various strategies for targeting NK cell receptors and NK cell receptor ligands using multivalent proteins to activate immunity against cancer. EXPERT OPINION NK cell receptors work in synergy to activate NK cell effector responses. Effective anti-cancer strategies will need to not only kill tumor cells but must also lead to the destruction of the tumor microenvironment. Immunotherapy based on NK cells and their receptors has the capacity to accomplish this through triggering lymphocyte cytotoxicity and cytokine production.
Collapse
Affiliation(s)
- Nicole C Smits
- a Department of Microbiology and Immunology and the Center for Synthetic Immunity , The Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| | - Tiffany A Coupet
- a Department of Microbiology and Immunology and the Center for Synthetic Immunity , The Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| | - Claire Godbersen
- a Department of Microbiology and Immunology and the Center for Synthetic Immunity , The Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| | - Charles L Sentman
- a Department of Microbiology and Immunology and the Center for Synthetic Immunity , The Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| |
Collapse
|
45
|
Fousek K, Ahmed N. The Evolution of T-cell Therapies for Solid Malignancies. Clin Cancer Res 2016; 21:3384-92. [PMID: 26240290 DOI: 10.1158/1078-0432.ccr-14-2675] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Primary resistant, recurrent, and relapsed solid tumors are often nonresponsive to conventional antineoplastic therapies. Moreover, in responsive tumors, the therapeutic-to-toxic range of these interventions remains quite narrow, such that side effects of therapy are substantial. Targeted therapies, such as adoptive T-cell transfer, not only spare normal tissues but also use alternative killing mechanisms to which the tumor cells are usually not immune. Adoptive T-cell transfer for solid tumors faces unique challenges because of the inherent heterogeneity of tumor parenchyma, the complexity of the tumor microenvironment, and tumor occurrence in areas with limited therapeutic accessibility. In this review, we examine the recent evolution of various T-cell-based immunotherapeutics, the mechanisms of action behind their antitumor activity, their increasing complexity, and the prospect of building on previous successes in the treatment of solid tumors.
Collapse
Affiliation(s)
- Kristen Fousek
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas. Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas. Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas. Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
| | - Nabil Ahmed
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas. Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas. Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas. Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
46
|
Yusibov V, Kushnir N, Streatfield SJ. Antibody Production in Plants and Green Algae. ANNUAL REVIEW OF PLANT BIOLOGY 2016; 67:669-701. [PMID: 26905655 DOI: 10.1146/annurev-arplant-043015-111812] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Monoclonal antibodies (mAbs) have a wide range of modern applications, including research, diagnostic, therapeutic, and industrial uses. Market demand for mAbs is high and continues to grow. Although mammalian systems, which currently dominate the biomanufacturing industry, produce effective and safe recombinant mAbs, they have a limited manufacturing capacity and high costs. Bacteria, yeast, and insect cell systems are highly scalable and cost effective but vary in their ability to produce appropriate posttranslationally modified mAbs. Plants and green algae are emerging as promising production platforms because of their time and cost efficiencies, scalability, lack of mammalian pathogens, and eukaryotic posttranslational protein modification machinery. So far, plant- and algae-derived mAbs have been produced predominantly as candidate therapeutics for infectious diseases and cancer. These candidates have been extensively evaluated in animal models, and some have shown efficacy in clinical trials. Here, we review ongoing efforts to advance the production of mAbs in plants and algae.
Collapse
Affiliation(s)
- Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware 19711; , ,
| | - Natasha Kushnir
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware 19711; , ,
| | | |
Collapse
|
47
|
Root AR, Cao W, Li B, LaPan P, Meade C, Sanford J, Jin M, O'Sullivan C, Cummins E, Lambert M, Sheehan AD, Ma W, Gatto S, Kerns K, Lam K, D'Antona AM, Zhu L, Brady WA, Benard S, King A, He T, Racie L, Arai M, Barrett D, Stochaj W, LaVallie ER, Apgar JR, Svenson K, Mosyak L, Yang Y, Chichili GR, Liu L, Li H, Burke S, Johnson S, Alderson R, Finlay WJJ, Lin L, Olland S, Somers W, Bonvini E, Gerber HP, May C, Moore PA, Tchistiakova L, Bloom L. Development of PF-06671008, a Highly Potent Anti-P-cadherin/Anti-CD3 Bispecific DART Molecule with Extended Half-Life for the Treatment of Cancer. Antibodies (Basel) 2016; 5:E6. [PMID: 31557987 PMCID: PMC6698862 DOI: 10.3390/antib5010006] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/14/2016] [Accepted: 02/17/2016] [Indexed: 01/13/2023] Open
Abstract
Bispecific antibodies offer a promising approach for the treatment of cancer but can be challenging to engineer and manufacture. Here we report the development of PF-06671008, an extended-half-life dual-affinity re-targeting (DART®) bispecific molecule against P-cadherin and CD3 that demonstrates antibody-like properties. Using phage display, we identified anti-P-cadherin single chain Fv (scFv) that were subsequently affinity-optimized to picomolar affinity using stringent phage selection strategies, resulting in low picomolar potency in cytotoxic T lymphocyte (CTL) killing assays in the DART format. The crystal structure of this disulfide-constrained diabody shows that it forms a novel compact structure with the two antigen binding sites separated from each other by approximately 30 Å and facing approximately 90° apart. We show here that introduction of the human Fc domain in PF-06671008 has produced a molecule with an extended half-life (-4.4 days in human FcRn knock-in mice), high stability (Tm1 > 68 °C), high expression (>1 g/L), and robust purification properties (highly pure heterodimer), all with minimal impact on potency. Finally, we demonstrate in vivo anti-tumor efficacy in a human colorectal/human peripheral blood mononuclear cell (PBMC) co-mix xenograft mouse model. These results suggest PF-06671008 is a promising new bispecific for the treatment of patients with solid tumors expressing P-cadherin.
Collapse
Affiliation(s)
- Adam R Root
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Wei Cao
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Bilian Li
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Peter LaPan
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Caryl Meade
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Jocelyn Sanford
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Macy Jin
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Cliona O'Sullivan
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Emma Cummins
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Matthew Lambert
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Alfredo D Sheehan
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Weijun Ma
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Scott Gatto
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Kelvin Kerns
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Khetemenee Lam
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Aaron M D'Antona
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Lily Zhu
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - William A Brady
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Susan Benard
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Amy King
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Tao He
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Lisa Racie
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Maya Arai
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Dianah Barrett
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Wayne Stochaj
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Edward R LaVallie
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - James R Apgar
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Kristine Svenson
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Lidia Mosyak
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Yinhua Yang
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | | | - Liqin Liu
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Hua Li
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Steve Burke
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Syd Johnson
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Ralph Alderson
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - William J J Finlay
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Laura Lin
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Stéphane Olland
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - William Somers
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Ezio Bonvini
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Hans-Peter Gerber
- Oncology Research Unit, Pfizer Inc., 401 N. Middletown Road, Pearl River, NY 10965, USA.
| | - Chad May
- Oncology Research Unit, Pfizer Inc., 401 N. Middletown Road, Pearl River, NY 10965, USA.
| | - Paul A Moore
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Lioudmila Tchistiakova
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Laird Bloom
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| |
Collapse
|
48
|
Postel-Vinay S, Aspeslagh S, Lanoy E, Robert C, Soria JC, Marabelle A. Challenges of phase 1 clinical trials evaluating immune checkpoint-targeted antibodies. Ann Oncol 2015; 27:214-24. [PMID: 26578728 DOI: 10.1093/annonc/mdv550] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/27/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Immunostimulatory monoclonal antibodies (imAbs) targeting immune checkpoint molecules are revolutionizing oncology not only regarding cancer therapeutics and clinical care, but also from a drug development point of view. A handful of first-generation molecules have been approved so far based on their tremendous efficacy, after an expedited development phase that has challenged most paradigms established in the era of conventional cytotoxic therapy and to some extent molecularly targeted agents. A huge wave of second-generation imAbs is just entering into phase 1 trials now, in monotherapy or in combination. In order to maximize their chances of success in early phase trials, and eventually for patients' benefit, their clinical development has to benefit from lessons learnt from previous imAbs phase 1 trials. MATERIALS AND METHODS We reviewed the early clinical development of anti-cytotoxic T-lymphocyte antigen 4 and anti-programmed death-1 receptor/ligand. Particularities of each agent, including safety, dose--toxicity and dose--efficacy relationships, scheduling, pharmacokinetics (PK), pharmacodynamics (PD), trial design, biomarkers, response assessment and overall drug development strategies, are described and challenged. RESULTS As opposed to conventional cytotoxic agents, dose of imAbs is not linearly associated with efficacy and toxicity. Therefore, the definition of a minimal immunologically active dose could be proposed. Traditional patient eligibility criteria might also be revisited as the toxicity profile and mechanism of toxicity--immune-related adverse events--are mostly known and their physiopathology somehow less unexpected than with molecularly targeted small molecules. Most challenging are the comprehensive investigation of complex PK and PD characteristics as well as the definition of patient selection biomarkers. Finally, the early focus on efficacy (and not only dose confirmation) in expansion cohorts challenges the traditional phase 1/2/3 drug development process. CONCLUSION Several drug development paradigms have been challenged by imAbs. Here, we discuss novel approaches for an efficient and successful drug development of these agents.
Collapse
Affiliation(s)
- S Postel-Vinay
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif
| | - S Aspeslagh
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif
| | - E Lanoy
- Biostatistics and Epidemiology Unit, Gustave-Roussy, Villejuif Inserm Unit U1018, CESP, Université Paris-Sud, Université Paris-Saclay, Villejuif
| | - C Robert
- Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif Department of Medical Oncology, Gustave Roussy, Villejuif
| | - J-C Soria
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif
| | - A Marabelle
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Inserm Unit U1015, Gustave Roussy, Villejuif, France
| |
Collapse
|
49
|
Rathi C, Meibohm B. Clinical pharmacology of bispecific antibody constructs. J Clin Pharmacol 2015; 55 Suppl 3:S21-8. [PMID: 25707960 DOI: 10.1002/jcph.445] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 12/09/2014] [Indexed: 12/25/2022]
Abstract
The confluence of rapid scientific advancements especially in protein engineering and recombinant technology, unmet medical needs, and commercial incentives have led to the development of the next generation of therapeutic proteins. Bispecific antibody constructs are one of the novel strategies that is being pursued, combining the ability to bind simultaneously to two distinct targets and the advantages of purpose-designed and optimized antibody-based scaffolds. Their pharmacokinetic and pharmacodynamic properties, including their immunogenic potential, are closely related to their structural features and ability to interact with disposition mechanisms of immunoglobulin molecules. Catumaxomab and blinatumomab are bispecific constructs that are approved for clinical use and have provided clinical pharmacology data for this novel class of therapeutics. This knowledgebase on the clinical behavior of bispecific therapeutic proteins is poised to rapidly evolve over the next few years with many development programs having entered the clinical development stage.
Collapse
Affiliation(s)
- Chetan Rathi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | | |
Collapse
|
50
|
Regan D, Guth A, Coy J, Dow S. Cancer immunotherapy in veterinary medicine: Current options and new developments. Vet J 2015; 207:20-28. [PMID: 26545847 DOI: 10.1016/j.tvjl.2015.10.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 09/24/2015] [Accepted: 10/04/2015] [Indexed: 12/12/2022]
Abstract
Excitement in the field of tumor immunotherapy is being driven by several remarkable breakthroughs in recent years. This review will cover recent advances in cancer immunotherapy, including the use of T cell checkpoint inhibitors, engineered T cells, cancer vaccines, and anti-B cell and T cell antibodies. Inhibition of T cell checkpoint molecules such as PD-1 and CTLA-4 using monoclonal antibodies has achieved notable success against advanced tumors in humans, including melanoma, renal cell carcinoma, and non-small cell lung cancer. Therapy with engineered T cells has also demonstrated remarkable tumor control and regression in human trials. Autologous cancer vaccines have recently demonstrated impressive prolongation of disease-free intervals and survival times in dogs with lymphoma. In addition, caninized monoclonal antibodies targeting CD20 and CD52 just recently received either full (CD20) or conditional (CD52) licensing by the United States Department of Agriculture for clinical use in the treatment of canine B-cell and T-cell lymphomas, respectively. Thus, immunotherapy for cancer is rapidly moving to the forefront of cancer treatment options in veterinary medicine as well as human medicine.
Collapse
Affiliation(s)
- Daniel Regan
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80523, USA
| | - Amanda Guth
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80523, USA
| | - Jonathan Coy
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80523, USA
| | - Steven Dow
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80523, USA.
| |
Collapse
|