101
|
Amatya C, Pegues MA, Lam N, Vanasse D, Geldres C, Choi S, Hewitt SM, Feldman SA, Kochenderfer JN. Development of CAR T Cells Expressing a Suicide Gene Plus a Chimeric Antigen Receptor Targeting Signaling Lymphocytic-Activation Molecule F7. Mol Ther 2020; 29:702-717. [PMID: 33129371 DOI: 10.1016/j.ymthe.2020.10.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/14/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptors (CARs) are fusion proteins that contain antigen-recognition domains and T cell signaling domains. Signaling lymphocytic-activation molecule F7 (SLAMF7) is a promising target for CAR T cell therapies of the plasma cell malignancy multiple myeloma (MM) because SLAMF7 is expressed by MM but not normal nonhematopoietic cells. We designed CARs targeting SLAMF7. We transduced human T cells with anti-SLAMF7 CARs containing either CD28 or 4-1BB costimulatory domains. T cells expressing CD28-containing CARs or 4-1BB-containing CARs recognized SLAMF7 in vitro. SLAMF7-specific cytokine release was higher for T cells expressing CARs with CD28 versus 4-1BB domains. In murine solid tumor and disseminated tumor models, anti-tumor activity of T cells was superior with CD28-containing CARs versus 4-1BB-containing CARs. Because of SLAMF7 expression on some normal leukocytes, especially natural killer cells that control certain viral infections, the inclusion of a suicide gene with an anti-SLAMF7 CAR is prudent. We designed a construct with a CD28-containing anti-SLAMF7 CAR and a suicide gene. The suicide gene encoded a dimerization domain fused to a caspase-9 domain. T cells expressing the anti-SLAMF7 CAR plus suicide-gene construct specifically recognized SLAMF7 in vitro and eliminated tumors from mice. T cells expressing this construct were eliminated on demand by administering the dimerizing agent AP1903 (rimiducid).
Collapse
Affiliation(s)
- Christina Amatya
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD 20892, USA
| | - Melissa A Pegues
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD 20892, USA
| | - Norris Lam
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD 20892, USA
| | - Danielle Vanasse
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD 20892, USA
| | - Claudia Geldres
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD 20892, USA
| | - Stephanie Choi
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD 20892, USA
| | - Stephen M Hewitt
- National Institutes of Health, National Cancer Institute Laboratory of Pathology, Bethesda, MD 20892, USA
| | | | - James N Kochenderfer
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD 20892, USA.
| |
Collapse
|
102
|
Yan W, Liu Z, Liu J, Xia Y, Hu K, Yu J. Application of Chimeric Antigen Receptor T Cells in the Treatment of Hematological Malignancies. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4241864. [PMID: 33062678 PMCID: PMC7547336 DOI: 10.1155/2020/4241864] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/17/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022]
Abstract
T cell immune protection plays a pivotal role in the treatment of patients with hematological malignancies. However, T cell exhaustion might lead to the possibility of immune escape of hematological malignancies. Adoptive cell therapy (ACT) with chimeric antigen receptor T (CAR-T) cells can restore the activity of exhausted T cell through reprogramming and is widely used in the treatment of relapsed/refractory (r/r) hematological malignancies. Of note, CD19, CD20, CD30, CD33, CD123, and CD269 as ideal targets have shown extraordinary potential for CAR-T cell therapy and other targets such as CD23 and SLAMF7 have brought promising future for clinical trials. However, CAR-T cells can also produce some adverse events after treatment of hematological malignancies, such as cytokine release syndrome (CRS), neurotoxicity, and on-target/off-tumor toxicity, which may cause systemic immune stress inflammation, destruction of the blood-brain barrier, and even normal tissue damage. In this review, we aim to summarize the composition of CAR-T cell and its application in the treatment of acute lymphocytic leukemia (ALL), chronic lymphocytic leukemia (CLL), non-Hodgkin's lymphoma (NHL), Hodgkin's lymphoma (HL), multiple myeloma (MM), and acute myeloid leukemia (AML). Moreover, we will review the disadvantages of CAR-T cell therapy and propose several comprehensive recommendations which might guide its development.
Collapse
Affiliation(s)
- Weiqi Yan
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Zhuojun Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Jia Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yuanshi Xia
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Kai Hu
- Department of Hematology, Beijing Boren Hospital, Beijing 100070, China
| | - Jian Yu
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| |
Collapse
|
103
|
Mikkilineni L, Kochenderfer JN. CAR T cell therapies for patients with multiple myeloma. Nat Rev Clin Oncol 2020; 18:71-84. [PMID: 32978608 DOI: 10.1038/s41571-020-0427-6] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2020] [Indexed: 12/21/2022]
Abstract
Despite several therapeutic advances over the past decade, multiple myeloma (MM) remains largely incurable, indicating a need for new treatment approaches. Chimeric antigen receptor (CAR) T cell therapy works by mechanisms distinct from those of other MM therapies and involves the modification of patient or donor T cells to target specific cell-surface antigens. B cell maturation antigen (BCMA) is expressed only on plasma cells, a small subset of B cells and MM cells, which makes it a suitable target antigen for such therapies. At the time of writing, data from >20 clinical trials involving anti-BCMA CAR T cells have demonstrated that patients with relapsed and/or refractory MM can achieve objective responses. These early investigations have been instrumental in demonstrating short-term safety and efficacy; however, most patients do not have disease remission lasting >18 months. Attempts to reduce or delay the onset of relapsed disease are underway and include identifying additional CAR T cell target antigens and methods of enhancing BCMA expression on MM cells. Engineering CAR T cells to enhance both the activity and safety of treatment continues to be a promising avenue for improvement. In this Review we summarize data from clinical trials that have been carried out to date, describe novel antigens that could be targeted in the future, and highlight potential future innovations that could enhance the efficacy and/or reduce the toxicities associated with CAR T cell therapies.
Collapse
Affiliation(s)
- Lekha Mikkilineni
- Surgery Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA.
| | | |
Collapse
|
104
|
Abstract
Despite considerable advances in treatment approaches in the past two decades, multiple myeloma remains an incurable disease. Treatments for myeloma continue to evolve with many emerging immunotherapies. The first immunotherapy used to treat hematologic cancers, including multiple myeloma, was an allogeneic stem cell transplant. In the mid-2000s, immunomodulatory drugs thalidomide, lenalidomide, and subsequently pomalidomide were proven to be effective in multiple myeloma and substantially improved survival. The next wave of immunotherapies for multiple myeloma included the monoclonal antibodies daratumumab and elotuzumab, which were approved by the Food and Drug Administration in 2015. Subsequently, a variety of immunotherapies have been developed for multiple myeloma, including chimeric antigen receptor T cells, bispecific antibodies, antibody drug conjugates, and checkpoint inhibitors. Many of these emerging treatments target the B cell maturation antigen, which is expressed on plasma cells, although several other novel receptors are also being studied. This review summarizes the evidence of these various immunotherapies, their mechanism of action, and data from clinical trials regarding the treatments' safety and efficacy.
Collapse
Affiliation(s)
- Urvi A Shah
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, USA
| | - Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, USA
| |
Collapse
|
105
|
Nicolini F, Bravaccini S, Mazza M, Gruszka AM, Tazzari M, MartÍn-Antonio B, Juan M, Ibrahim T, Maltoni R, Martinelli G, Cerchione C. CAR T cells targeting options in the fight against multiple myeloma. Panminerva Med 2020; 63:37-45. [PMID: 32955187 DOI: 10.23736/s0031-0808.20.04146-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
INTRODUCTION Multiple myeloma (MM) is a hematological malignancy in which patients present with bone marrow infiltration of clonal terminally-differentiated plasma cells. Monoclonal protein in the serum and/or urine is frequently detected. Over the past decade, important progress has been made in the comprehension of disease biology and treatment personalization. Much work has been put into the development of chimeric antigen receptor (CAR) gene-modified T-cell therapy thought to be a promising therapeutic option for pluritreated patients with refractory MM. EVIDENCE ACQUISITION We performed an analysis of clinical trials registered at the international repository clinicaltrials.gov using "CAR" OR "CAR T" AND "multiple myeloma" as search terms to understand what were the antigens targeted by CAR T strategies and what was the trade-off of their exploitation. The search retrieved a list of 103 trials that was manually filtered to eliminate follow-up and observational or not-pertinent trials. EVIDENCE SYNTHESIS Most studies employed anti-BCMA targeting either alone (62/94; 66%), or in combination with a second target (12/94; 13%). The second target most studied was SLAMF7 (CD319) explored by 4/94 (4%) clinical trials. Other antigens investigated and described here include: CD44v6, CD38, CD138, MUC1, CD56, CD19, Igk light chain, Lewis Y, CD229 and GPRC5D. CONCLUSIONS Targeting an appropriate antigen(s) is the key to both safety and efficacy of CAR T approaches in MM as there is dearth of tumor-specific antigens. Most antigens tested are merely enriched on MM cells. Working with tumor-enriched antigens requires careful assessment of the balance between harm (toxicity) and benefit (disease eradication) to the patient. This review provides an up-to-date overview of the avenues that are being explored.
Collapse
Affiliation(s)
- Fabio Nicolini
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Sara Bravaccini
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Massimiliano Mazza
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy -
| | - Alicja M Gruszka
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Milan, Italy
| | - Marcella Tazzari
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Beatriz MartÍn-Antonio
- August Pi Biomedical Research Institute, Sunyer Hospital, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Manel Juan
- August Pi Biomedical Research Institute, Sunyer Hospital, Clinic of Immunology, Barcelona, Spain
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Roberta Maltoni
- Department of Medical Oncology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Giovanni Martinelli
- IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Claudio Cerchione
- IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| |
Collapse
|
106
|
Chimeric Antigen Receptor T-cell Therapy for Multiple Myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 21:21-34. [PMID: 33046423 DOI: 10.1016/j.clml.2020.08.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 12/17/2022]
Abstract
Relapsed/refractory multiple myeloma (MM) remains a significant clinical challenge, despite a wide array of approved therapeutic agents. Immunotherapy offers an advantage in this setting. Chimeric antigen receptor (CAR) modified T-cells have transformed care for patients with hematologic malignancies. CAR-T cells targeting CD-19 B-cell lymphoma cells have shown prominent activity in lymphoma and acute lymphoblastic leukemia. Recently, the CAR-T cell platform for MM demonstrated therapeutic benefit. Hence, it is rapidly progressing. The most commonly tested target for MM is the B-cell maturation antigen. Complexities involved in the generation and use of CAR-T cells for MM include the identification of appropriate target antigens that are specific, and tumor type restricted, in addition to the optimization of CAR constructs to mitigate toxicities including cytokine release syndrome. CAR-T cells hold immense promise as a therapeutic modality for the treatment of MM. In this article, we provide an updated review of clinical trials of MM-specific CAR-T cells.
Collapse
|
107
|
Amberger M, Ivics Z. Latest Advances for the Sleeping Beauty Transposon System: 23 Years of Insomnia but Prettier than Ever: Refinement and Recent Innovations of the Sleeping Beauty Transposon System Enabling Novel, Nonviral Genetic Engineering Applications. Bioessays 2020; 42:e2000136. [PMID: 32939778 DOI: 10.1002/bies.202000136] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/29/2020] [Indexed: 12/13/2022]
Abstract
The Sleeping Beauty transposon system is a nonviral DNA transfer tool capable of efficiently mediating transposition-based, stable integration of DNA sequences of choice into eukaryotic genomes. Continuous refinements of the system, including the emergence of hyperactive transposase mutants and novel approaches in vectorology, greatly improve upon transposition efficiency rivaling viral-vector-based methods for stable gene insertion. Current developments, such as reversible transgenesis and proof-of-concept RNA-guided transposition, further expand on possible applications in the future. In addition, innate advantages such as lack of preferential integration into genes reduce insertional mutagenesis-related safety concerns while comparably low manufacturing costs enable widespread implementation. Accordingly, the system is recognized as a powerful and versatile tool for genetic engineering and is playing a central role in an ever-expanding number of gene and cell therapy clinical trials with the potential to become a key technology to meet the growing demand for advanced therapy medicinal products.
Collapse
Affiliation(s)
- Maximilian Amberger
- Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, D-63225, Germany
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, D-63225, Germany
| |
Collapse
|
108
|
Rational design of a trimeric APRIL-based CAR-binding domain enables efficient targeting of multiple myeloma. Blood Adv 2020; 3:3248-3260. [PMID: 31698455 DOI: 10.1182/bloodadvances.2019000703] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/21/2019] [Indexed: 01/20/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells (CARTs) have shown tremendous potential for the treatment of certain B-cell malignancies, including patients with relapsed/refractory multiple myeloma (MM). Targeting the B-cell maturation antigen (BCMA) has produced the most promising results for CART therapy of MM to date, but not all remissions are sustained. Emergence of BCMA escape variants has been reported under the selective pressure of monospecific anti-BCMA CART treatment. Thus, there is a clinical need for continuous improvement of CART therapies for MM. Here, we show that a novel trimeric APRIL (a proliferation-inducing ligand)-based CAR efficiently targets both BCMA+ and BCMA- MM. Modeled after the natural ligand-receptor pair, APRIL-based CARs allow for bispecific targeting of the MM-associated antigens BCMA and transmembrane activator and CAML interactor (TACI). However, natural ligands as CAR antigen-binding domains may require further engineering to promote optimal binding and multimerization to adequately trigger T-cell activation. We found that using a trimeric rather than a monomeric APRIL format as the antigen-binding domain enhanced binding to BCMA and TACI and CART activity against MM in vitro and in vivo. Dual-specific, trimeric APRIL-based CAR are a promising therapeutic approach for MM with potential for preventing and treating BCMA escape.
Collapse
|
109
|
Chimeric antigen receptor T cell therapy in multiple myeloma: promise and challenges. Bone Marrow Transplant 2020; 56:9-19. [PMID: 32770147 DOI: 10.1038/s41409-020-01023-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/29/2020] [Indexed: 11/09/2022]
Abstract
Despite a sea change in the therapeutic landscape, multiple myeloma (MM), a cancer of antibody producing plasma cells, remains incurable requiring continued intervention for disease control. In this context, chimeric antigen receptor (CAR) T cell therapy has emerged as a promising immunotherapeutic approach with unprecedented results in heavily treated relapsed and/or refractory MM patients. Although B cell maturation antigen (BCMA) is the current lead target for CAR-T cell therapy in MM, several other antigenic targets are also being investigated. Relapses, however, are inevitable in spite of the promising early responses, and may be mediated by antigenic modulation, poor persistence and "immunostat" in tumor microenvironment. Akin to multi-agent chemotherapy, multi-targeted CAR-T antigens and combinatorial approaches are underway to overcome the resistance mechanisms. Further, CAR-T specific toxicity concerns such as cytokine release syndrome and neurotoxicity, as well as manufacturing time lag are other key challenges. Allogeneic CAR that offers "off-the-shelf" options, and mRNA transfected CAR are being developed to mitigate the access and safety issues. In this review we provide the comprehensive review of the most current clinical trial data for CAR-T in myeloma, challenges associated with this therapy and discuss its future in myeloma therapeutics.
Collapse
|
110
|
van der Schans JJ, van de Donk NWCJ, Mutis T. Dual Targeting to Overcome Current Challenges in Multiple Myeloma CAR T-Cell Treatment. Front Oncol 2020; 10:1362. [PMID: 32850436 PMCID: PMC7419675 DOI: 10.3389/fonc.2020.01362] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022] Open
Abstract
In the era of highly promising novel targeted-immunotherapy strategies for multiple myeloma (MM), the first series of clinical trials with CAR T-cells targeting the plasma cell-specific B-cell maturation antigen (BCMA) have shown excellent response rates. In the long-term, however, MM appears to escape the therapy likely due to initial low and heterogeneous expression or downregulation of BCMA expression. Several other molecules targeted by CAR T-cells in MM are expressed at high levels on MM cells, but many of these attractive targets are also expressed on various, sometimes vital non-malignant cells, posing major risks for on-target, off-tumor side effects. CAR T-cell therapy for MM therefore faces two urgent challenges: (i) improving the efficacy of BCMA CAR T-cells and (ii) establishing a MM-selectivity even when CAR T-cells are directed against not entirely MM-specific target antigens. In this review, we will outline the current attempts to tackle these challenges, with a specific focus on how dual CAR targeting might be applied to tackle both issues.
Collapse
Affiliation(s)
| | | | - Tuna Mutis
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
111
|
Targeting Multiple Myeloma through the Biology of Long-Lived Plasma Cells. Cancers (Basel) 2020; 12:cancers12082117. [PMID: 32751699 PMCID: PMC7466116 DOI: 10.3390/cancers12082117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/17/2020] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of terminally differentiated bone marrow (BM) resident B lymphocytes known as plasma cells (PC). PC that reside in the bone marrow include a distinct population of long-lived plasma cells (LLPC) that have the capacity to live for very long periods of time (decades in the human population). LLPC biology is critical for understanding MM disease induction and progression because MM shares many of the same extrinsic and intrinsic survival programs as LLPC. Extrinsic survival signals required for LLPC survival include soluble factors and cellular partners in the bone marrow microenvironment. Intrinsic programs that enhance cellular fidelity are also required for LLPC survival including increased autophagy, metabolic fitness, the unfolded protein response (UPR), and enhanced responsiveness to endoplasmic reticulum (ER) stress. Targeting LLPC cell survival mechanisms have led to standard of care treatments for MM including proteasome inhibition (Bortezomib), steroids (Dexamethasone), and immunomodulatory drugs (Lenalidomide). MM patients that relapse often do so by circumventing LLPC survival pathways targeted by treatment. Understanding the mechanisms by which LLPC are able to survive can allow us insight into the treatment of MM, which allows for the enhancement of therapeutic strategies in MM both at diagnosis and upon patient relapse.
Collapse
|
112
|
Rodríguez-Lobato LG, Ganzetti M, Fernández de Larrea C, Hudecek M, Einsele H, Danhof S. CAR T-Cells in Multiple Myeloma: State of the Art and Future Directions. Front Oncol 2020; 10:1243. [PMID: 32850376 PMCID: PMC7399644 DOI: 10.3389/fonc.2020.01243] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/16/2020] [Indexed: 01/24/2023] Open
Abstract
Despite recent therapeutic advances, the prognosis of multiple myeloma (MM) patients remains poor. Thus, new strategies to improve outcomes are imperative. Chimeric antigen receptor (CAR) T-cell therapy has changed the treatment landscape of B-cell malignancies, providing a potentially curative option for patients who are refractory to standard treatment. Long-term remissions achieved in patients with acute lymphoblastic leukemia and Non-Hodgkin Lymphoma encouraged its further development in MM. B-cell maturation antigen (BCMA)-targeted CAR T-cells have established outstanding results in heavily pre-treated patients. However, several other antigens such as SLAMF7 and CD44v6 are currently under investigation with promising results. Idecabtagene vicleucel is expected to be approved soon for clinical use. Unfortunately, relapses after CAR T-cell infusion have been reported. Hence, understanding the underlying mechanisms of resistance is essential to promote prevention strategies and to enhance CAR T-cell efficacy. In this review we provide an update of the most recent clinical and pre-clinical data and we elucidate both, the potential and the challenges of CAR T-cell therapy in the future.
Collapse
Affiliation(s)
- Luis Gerardo Rodríguez-Lobato
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maya Ganzetti
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carlos Fernández de Larrea
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Michael Hudecek
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Sophia Danhof
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
113
|
Clinical data, limitations and perspectives on chimeric antigen receptor T-cell therapy in multiple myeloma. Curr Opin Oncol 2020; 32:418-426. [DOI: 10.1097/cco.0000000000000667] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
114
|
Zhu S, Chen Y, Lao J, Wu C, Zhan X, Wu Y, Shang Y, Zou Z, Zhou J, Ji X, Huang X, Shi X, Wu M. Signaling Lymphocytic Activation Molecule Family-7 Alleviates Corneal Inflammation by Promoting M2 Polarization. J Infect Dis 2020; 223:854-865. [PMID: 32702113 DOI: 10.1093/infdis/jiaa445] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/11/2020] [Accepted: 07/17/2020] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Signaling lymphocytic activation molecule family-7 (SLAMF7) functions as an immune checkpoint molecule on macrophages in antitumor immunity. However, its role in bacterial infection remains largely unknown. METHODS Bone marrow-derived macrophages (BMDMs) isolated from wild-type (WT) or SLAMF7 knockout (KO) mice were infected with bacteria or treated with lipopolysaccharide/interferon-γ to investigate the expression and function of SLAMF7 in macrophage polarization. A Pseudomonas aeruginosa keratitis murine model was established to explore the effect of SLAMF7 on P. aeruginosa keratitis using WT vs SLAMF7 KO mice, or recombinant SLAMF7 vs phosphate-buffered saline-treated mice, respectively. RESULTS SLAMF7 expression was enhanced on M1-polarized or bacterial-infected macrophages, and infiltrating macrophages in P. aeruginosa-infected mouse corneas. SLAMF7 promoted M2 polarization by inducing STAT6 activation. In vivo data showed that SLAMF7 KO aggravated, while treatment with recombinant SLAMF7 alleviated, corneal inflammation and disease severity. In addition, blockage of M2 polarization by Arg-1 inhibitor abrogated the effect of recombinant SLAMF7 in disease progression. CONCLUSIONS SLAMF7 expression in macrophages was induced upon M1 polarization or bacterial infection and alleviated corneal inflammation and disease progression of P. aeruginosa keratitis by promoting M2 polarization. These findings may provide a potential strategy for the treatment of P. aeruginosa keratitis.
Collapse
Affiliation(s)
- Siying Zhu
- Program of Infection and Immunology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Yu Chen
- Program of Infection and Immunology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Juanfeng Lao
- Program of Infection and Immunology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Chenglin Wu
- Organ Transplantation Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoxia Zhan
- Department of Laboratory Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongjian Wu
- Program of Infection and Immunology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Yuqi Shang
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Zhengyu Zou
- Program of Infection and Immunology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Jinyu Zhou
- Program of Infection and Immunology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Xiang Ji
- Program of Infection and Immunology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Xi Huang
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Xiaomin Shi
- Organ Transplantation Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minhao Wu
- Program of Infection and Immunology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Guangdong Engineering and Technology Research Center for Disease-Model Animals, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
115
|
Shah N, Aiello J, Avigan DE, Berdeja JG, Borrello IM, Chari A, Cohen AD, Ganapathi K, Gray L, Green D, Krishnan A, Lin Y, Manasanch E, Munshi NC, Nooka AK, Rapoport AP, Smith EL, Vij R, Dhodapkar M. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of multiple myeloma. J Immunother Cancer 2020; 8:e000734. [PMID: 32661116 PMCID: PMC7359060 DOI: 10.1136/jitc-2020-000734] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2020] [Indexed: 12/24/2022] Open
Abstract
Outcomes in multiple myeloma (MM) have improved dramatically in the last two decades with the advent of novel therapies including immunomodulatory agents (IMiDs), proteasome inhibitors and monoclonal antibodies. In recent years, immunotherapy for the treatment of MM has advanced rapidly, with the approval of new targeted agents and monoclonal antibodies directed against myeloma cell-surface antigens, as well as maturing data from late stage trials of chimeric antigen receptor CAR T cells. Therapies that engage the immune system to treat myeloma offer significant clinical benefits with durable responses and manageable toxicity profiles, however, the appropriate use of these immunotherapy agents can present unique challenges for practicing physicians. Therefore, the Society for Immunotherapy of Cancer convened an expert panel, which met to consider the current role of approved and emerging immunotherapy agents in MM and provide guidance to the oncology community by developing consensus recommendations. As immunotherapy evolves as a therapeutic option for the treatment of MM, these guidelines will be updated.
Collapse
Affiliation(s)
- Nina Shah
- Division of Hematology-Oncology, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jack Aiello
- Patient Empowerment Network, San Jose, California, USA
| | - David E Avigan
- Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jesus G Berdeja
- Department of Medicine, Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | - Ivan M Borrello
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center of Johns Hopkins, Baltimore, Maryland, USA
| | - Ajai Chari
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adam D Cohen
- Department of Medicine, Abramson Cancer Center at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Karthik Ganapathi
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Lissa Gray
- University of California San Francisco, San Francisco, CA, USA
| | - Damian Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Amrita Krishnan
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Multiple Myeloma Center for Clinical Research, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Yi Lin
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elisabet Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nikhil C Munshi
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Ajay K Nooka
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia, USA
| | - Aaron P Rapoport
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Eric L Smith
- Myeloma Service and Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ravi Vij
- Division of Medical Oncology, Siteman Cancer Center, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Madhav Dhodapkar
- School of Medicine, Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
116
|
Development of CAR-T cell therapies for multiple myeloma. Leukemia 2020; 34:2317-2332. [PMID: 32572190 DOI: 10.1038/s41375-020-0930-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/29/2020] [Accepted: 06/12/2020] [Indexed: 12/15/2022]
Abstract
Currently available data on chimeric antigen receptor (CAR)-T cell therapy has demonstrated efficacy and manageable toxicity in heavily pretreated multiple myeloma (MM) patients. The CAR-T field in MM is rapidly evolving with >50 currently ongoing clinical trials across all phases, different CAR-T design, or targets. Most of the CAR-T trials are performed in China and the United States, while European centers organize or participate in only a small fraction of current clinical investigations. Autologous CAR-T cell therapy against B cell maturation antigen shows the best evidence of efficacy so far but main issues remain to be addressed: duration of response, longer follow-up, prolonged cytopenia, patients who may benefit the most such as those with extramedullary disease, outcome prediction, and the integration of CAR-T cell therapy within the MM treatment paradigm. Other promising targets are, i.a.,: CD38, SLAMF7/CS1, or GPRC5D. Although no product has been approved to date, cost and production time for autologous products are expected to be the main obstacles for broad use, for which reason allogeneic CAR-T cells are currently explored. However, the inherent risk of graft-versus-host disease requires additional modification which still need to be validated. This review aims to present the current status of CAR-T cell therapy in MM with an overview on current targets, designs, and stages of CAR-T cell development. Main challenges to CAR-T cell therapy will be highlighted as well as strategies to structurally improve the CAR-T cell product, and thereby its efficacy and safety. The need for comparability of the most promising therapies will be emphasized to balance risks and benefits in an evidence-based but personalized approach to further improve outcome of patients with MM.
Collapse
|
117
|
Feng D, Sun J. Overview of anti-BCMA CAR-T immunotherapy for multiple myeloma and relapsed/refractory multiple myeloma. Scand J Immunol 2020; 92:e12910. [PMID: 32471019 DOI: 10.1111/sji.12910] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/23/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) is a haematological malignancy caused by malignant proliferation of plasma cells in bone marrow. In recent years, MM patients are commonly treated with chemotherapy, autologous stem cell transplantation, protease inhibitors, immunomodulatory drugs and monoclonal antibodies, however most patients eventually relapse. Therefore, more effective therapies are highly needed. Anti-BCMA CAR-T therapy, a novel and efficacious method for treating MM and relapsed/refractory multiple myeloma (RRMM), has been designed and applied in clinics. The CAR-T can specifically recognize the targeted molecule B cell maturation antigen (BCMA) and kill MM cells expressing BCMA and several clinical trials have revealed high response rates in the therapy. Herein, we summarize the developments, the current design and clinical trials, the side effects of anti-BCMA CAR-T therapy and comparison of it with other CAR-T therapies.
Collapse
Affiliation(s)
- Deming Feng
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Jian Sun
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
118
|
Cho SF, Lin L, Xing L, Li Y, Yu T, Anderson KC, Tai YT. BCMA-Targeting Therapy: Driving a New Era of Immunotherapy in Multiple Myeloma. Cancers (Basel) 2020; 12:E1473. [PMID: 32516895 PMCID: PMC7352710 DOI: 10.3390/cancers12061473] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
The treatment of multiple myeloma (MM) has entered into a new era of immunotherapy. Novel immunotherapies will significantly improve patient outcome via simultaneously targeting malignant plasma cell (PC) and reversing immunocompromised bone marrow (BM) microenvironment. B-cell maturation antigen (BCMA), selectively expressed in PCs and a key receptor for A proliferation-inducing ligand (APRIL), is highly expressed in MM cells from patients at all stages. The APRIL/BCMA signal cascades promote the survival and drug resistance of MM cells and further modulate immunosuppressive BM milieu. Impressively, anti-BCMA immunotherapeutic reagents, including chimeric antigen receptor (CAR), antibody-drug conjugate (ADC) and bispecific T cell engager (BiTE) have all shown high response rates in their first clinical trials in relapse and refractory patients with very limited treatment options. These results rapidly inspired numerous development of next-generation anti-BCMA biotherapeutics, i.e., bispecific molecule, bispecific or trispecific antibodies, a novel form of CAR T/NK cells and T Cell Antigen Coupler (TAC) receptors, antibody-coupled T cell receptor (ACTR) as well as a cancer vaccine. We here highlight seminal preclinical and clinical studies on novel BCMA-based immunotherapies as effective monotherapy and discuss their potential in combination with current anti-MM and novel checkpoint drugs in earlier disease stages to further achieve durable responses in patients.
Collapse
Affiliation(s)
- Shih-Feng Cho
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Liang Lin
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| | - Lijie Xing
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| | - Yuyin Li
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| | - Tengteng Yu
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| |
Collapse
|
119
|
Magnani CF, Tettamanti S, Alberti G, Pisani I, Biondi A, Serafini M, Gaipa G. Transposon-Based CAR T Cells in Acute Leukemias: Where are We Going? Cells 2020; 9:cells9061337. [PMID: 32471151 PMCID: PMC7349235 DOI: 10.3390/cells9061337] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 02/07/2023] Open
Abstract
Chimeric Antigen Receptor (CAR) T-cell therapy has become a new therapeutic reality for refractory and relapsed leukemia patients and is also emerging as a potential therapeutic option in solid tumors. Viral vector-based CAR T-cells initially drove these successful efforts; however, high costs and cumbersome manufacturing processes have limited the widespread clinical implementation of CAR T-cell therapy. Here we will discuss the state of the art of the transposon-based gene transfer and its application in CAR T immunotherapy, specifically focusing on the Sleeping Beauty (SB) transposon system, as a valid cost-effective and safe option as compared to the viral vector-based systems. A general overview of SB transposon system applications will be provided, with an update of major developments, current clinical trials achievements and future perspectives exploiting SB for CAR T-cell engineering. After the first clinical successes achieved in the context of B-cell neoplasms, we are now facing a new era and it is paramount to advance gene transfer technology to fully exploit the potential of CAR T-cells towards next-generation immunotherapy.
Collapse
|
120
|
Paving the Way toward Successful Multiple Myeloma Treatment: Chimeric Antigen Receptor T-Cell Therapy. Cells 2020; 9:cells9040983. [PMID: 32316105 PMCID: PMC7226998 DOI: 10.3390/cells9040983] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 12/19/2022] Open
Abstract
Despite the significant progress of modern anticancer therapies, multiple myeloma (MM) is still incurable for the majority of patients. Following almost three decades of development, chimeric antigen receptor (CAR) T-cell therapy now has the opportunity to revolutionize the treatment landscape and meet the unmet clinical need. However, there are still several major hurdles to overcome. Here we discuss the recent advances of CAR T-cell therapy for MM with an emphasis on future directions and possible risks. Currently, CAR T-cell therapy for MM is at the first stage of clinical studies, and most studies have focused on CAR T cells targeting B cell maturation antigen (BCMA), but other antigens such as cluster of differentiation 138 (CD138, syndecan-1) are also being evaluated. Although this therapy is associated with side effects, such as cytokine release syndrome and neurotoxicity, and relapses have been observed, the benefit–risk balance and huge potential drive the ongoing clinical progress. To fulfill the promise of recent clinical trial success and maximize the potential of CAR T, future efforts should focus on the reduction of side effects, novel targeted antigens, combinatorial uses of different types of CAR T, and development of CAR T cells targeting more than one antigen.
Collapse
|
121
|
Beck JD, Birtel M, Haefner E, Keil IS, Reidenbach D, Salomon N, Yildiz IG, Diken M. CIMT 2019: report on the 17th Annual Meeting of the Association for Cancer Immunotherapy. Hum Vaccin Immunother 2020; 16:808-815. [PMID: 31584850 PMCID: PMC7227697 DOI: 10.1080/21645515.2019.1675459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Jan David Beck
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Matthias Birtel
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Erik Haefner
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Isabell Sofia Keil
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Daniel Reidenbach
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Nadja Salomon
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Ikra Gizem Yildiz
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Mustafa Diken
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| |
Collapse
|
122
|
Abstract
Multiple myeloma (MM), a bone marrow-resident hematological malignancy of plasma cells, has remained largely incurable despite dramatic improvements in patient outcomes in the era of myeloma-targeted and immunomodulatory agents. It has recently become clear that T cells from MM patients are able to recognize and eliminate myeloma, although this is subverted in the majority of patients who eventually succumb to progressive disease. T cell exhaustion and a suppressive bone marrow microenvironment have been implicated in disease progression, and once these are established, immunotherapy appears largely ineffective. Autologous stem cell transplantation (ASCT) is a standard of care in eligible patients and results in immune effects beyond cytoreduction, including lymphodepletion, T cell priming via immunogenic cell death, and inflammation; all occur within the context of a disrupted bone marrow microenvironment. Recent studies suggest that ASCT reestablishes immune equilibrium and thus represents a logical platform in which to intervene to prevent immune escape. New immunotherapies based on checkpoint inhibition targeting the immune receptor TIGIT and the deletion of suppressive myeloid populations appear attractive, particularly after ASCT. Finally, the immunologically favorable environment created after ASCT may also represent an opportunity for approaches utilizing bispecific antibodies or chimeric antigen receptor T cells.
Collapse
Affiliation(s)
- Simone A. Minnie
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Geoffrey R. Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Division of Medical Oncology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
123
|
Prommersberger S, Hudecek M, Nerreter T. Antibody-Based CAR T Cells Produced by Lentiviral Transduction. ACTA ACUST UNITED AC 2020; 128:e93. [PMID: 32150338 DOI: 10.1002/cpim.93] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
One promising approach to treat hematologic malignancies is the usage of patient-derived CAR T cells. There are continuous efforts to improve the function of these cells, to optimize their receptor, and to use them for the treatment of additional types of cancer and especially solid tumors. In this protocol, an easy and reliable approach for CAR T cell generation is described. T cells are first isolated from peripheral blood (here: leukoreduction system chambers) and afterwards activated for one day with anti-CD3/CD28 Dynabeads. The gene transfer is performed by lentiviral transduction and gene transfer rate can be verified by flowcytometric analysis. Six days after transduction, the stimulatory Dynabeads are removed. T cells are cultured in interleukin-2 conditioned medium for several days for expansion. There is an option to expand CAR T cells further by co-incubation with irradiated, antigen-expressing feeder cell lines. The CAR T cells are ready to use after 10 (without feeder cell expansion) to 24 days (with feeder cell expansion). © 2020 The Authors. Basic Protocol: Generation of CAR T cells by lentiviral transduction.
Collapse
|
124
|
Potent CAR-T cells engineered with Sleeping Beauty transposon vectors display a central memory phenotype. Gene Ther 2020; 28:3-5. [PMID: 32139891 DOI: 10.1038/s41434-020-0138-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 12/19/2022]
|
125
|
Shah UA, Mailankody S. CAR T and CAR NK cells in multiple myeloma: Expanding the targets. Best Pract Res Clin Haematol 2020; 33:101141. [PMID: 32139020 PMCID: PMC7137578 DOI: 10.1016/j.beha.2020.101141] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/09/2020] [Indexed: 12/21/2022]
Abstract
Multiple myeloma (MM) is a haematologic malignancy with significant improvements in the overall survival over the last decade. However, patients still relapse and die due to a lack of treatment options. Ultimately, novel therapies with the potential for long term remissions are needed for patients with advanced MM. Research efforts for such immune therapies were not successful until recently when the first immunotherapies for MM were approved in 2015 and many more are under development. In this review, we focus on adoptive cell therapies including CAR T-cell and CAR NK-cell therapies for patients with MM. We will provide an update on clinical and translational advances with a focus on results from ongoing clinical trials with BCMA targeted cellular therapies and the development of other novel targets, changes in the manufacturing process, trials focusing on earlier lines of therapy and combinations with other therapies as well as off the shelf products.
Collapse
Affiliation(s)
- Urvi A Shah
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Sham Mailankody
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
126
|
Ashmore-Harris C, Fruhwirth GO. The clinical potential of gene editing as a tool to engineer cell-based therapeutics. Clin Transl Med 2020; 9:15. [PMID: 32034584 PMCID: PMC7007464 DOI: 10.1186/s40169-020-0268-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
The clinical application of ex vivo gene edited cell therapies first began a decade ago with zinc finger nuclease editing of autologous CD4+ T-cells. Editing aimed to disrupt expression of the human immunodeficiency virus co-receptor gene CCR5, with the goal of yielding cells resistant to viral entry, prior to re-infusion into the patient. Since then the field has substantially evolved with the arrival of the new editing technologies transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeats (CRISPR), and the potential benefits of gene editing in the arenas of immuno-oncology and blood disorders were quickly recognised. As the breadth of cell therapies available clinically continues to rise there is growing interest in allogeneic and off-the-shelf approaches and multiplex editing strategies are increasingly employed. We review here the latest clinical trials utilising these editing technologies and consider the applications on the horizon.
Collapse
Affiliation(s)
- Candice Ashmore-Harris
- Imaging Therapy and Cancer Group, Dept of Imaging Chemistry & Biology, School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London (KCL), London, SE1 7EH, UK
- Centre for Stem Cells & Regenerative Medicine, School of Basic and Medical Biosciences, Guy's Hospital, KCL, London, SE1 9RT, UK
| | - Gilbert O Fruhwirth
- Imaging Therapy and Cancer Group, Dept of Imaging Chemistry & Biology, School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London (KCL), London, SE1 7EH, UK.
| |
Collapse
|
127
|
CD229 CAR T cells eliminate multiple myeloma and tumor propagating cells without fratricide. Nat Commun 2020; 11:798. [PMID: 32034142 PMCID: PMC7005855 DOI: 10.1038/s41467-020-14619-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/21/2020] [Indexed: 01/17/2023] Open
Abstract
Multiple myeloma (MM) is a plasma cell malignancy and most patients eventually succumb to the disease. Chimeric antigen receptor (CAR) T cells targeting B-Cell Maturation Antigen (BCMA) on MM cells have shown high-response rates, but limited durability. CD229/LY9 is a cell surface receptor present on B and T lymphocytes that is universally and strongly expressed on MM plasma cells. Here, we develop CD229 CAR T cells that are highly active in vitro and in vivo against MM plasma cells, memory B cells, and MM-propagating cells. We do not observe fratricide during CD229 CAR T cell production, as CD229 is downregulated in T cells during activation. In addition, while CD229 CAR T cells target normal CD229high T cells, they spare functional CD229neg/low T cells. These findings indicate that CD229 CAR T cells may be an effective treatment for patients with MM.
Collapse
|
128
|
Jacoby E, Shahani SA, Shah NN. Updates on CAR T-cell therapy in B-cell malignancies. Immunol Rev 2020; 290:39-59. [PMID: 31355492 DOI: 10.1111/imr.12774] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 05/09/2019] [Indexed: 12/22/2022]
Abstract
By increasing disease-free survival and offering the potential for long-term cure, chimeric antigen receptor (CAR) T-cell therapy has dramatically expanded therapeutic options among those with high-risk B-cell malignancies. As CAR T-cell utilization evolves however, novel challenges are generated. These include determining how to optimally integrate CAR T cells into standard of care and overcoming mechanisms of resistance to CAR T-cell therapy, such as evolutionary stress induced on cancer cells leading to immunophenotypic changes that allow leukemia to evade this targeted therapy. Compounding these challenges are the limited ability to determine differences between various CAR T-cell constructs, understanding the generalizability of trial outcomes from multiple sites utilizing unique CAR manufacturing strategies, and comparing distinct criteria for toxicity grading while defining optimal management. Additionally, as understanding of CAR behavior in humans has developed, strategies have appropriately evolved to proactively mitigate toxicities. These challenges offer complimentary insights and guide next steps to enhance the efficacy of this novel therapeutic modality. With a focus on B-cell malignancies as the paradigm for effective CAR T-cell therapy, this review describes advances in the field as well as current challenges and future directions.
Collapse
Affiliation(s)
- Elad Jacoby
- Division of Pediatric Hematology, Oncology and BMT, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shilpa A Shahani
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
129
|
Huang H, Wu HW, Hu YX. Current advances in chimeric antigen receptor T-cell therapy for refractory/relapsed multiple myeloma. J Zhejiang Univ Sci B 2020; 21:29-41. [PMID: 31898440 PMCID: PMC6964993 DOI: 10.1631/jzus.b1900351] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/30/2019] [Indexed: 01/07/2023]
Abstract
Multiple myeloma (MM), considered an incurable hematological malignancy, is characterized by its clonal evolution of malignant plasma cells. Although the application of autologous stem cell transplantation (ASCT) and the introduction of novel agents such as immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs) have doubled the median overall survival to eight years, relapsed and refractory diseases are still frequent events in the course of MM. To achieve a durable and deep remission, immunotherapy modalities have been developed for relapsed/refractory multiple myeloma (RRMM). Among these approaches, chimeric antigen receptor (CAR) T-cell therapy is the most promising star, based on the results of previous success in B-cell neoplasms. In this immunotherapy, autologous T cells are engineered to express an artificial receptor which targets a tumor-associated antigen and initiates the T-cell killing procedure. Tisagenlecleucel and Axicabtagene, targeting the CD19 antigen, are the two pacesetters of CAR T-cell products. They were approved by the US Food and Drug Administration (FDA) in 2017 for the treatment of acute lymphocytic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL). Their development enabled unparalleled efficacy in combating hematopoietic neoplasms. In this review article, we summarize six promising candidate antigens in MM that can be targeted by CARs and discuss some noteworthy studies of the safety profile of current CAR T-cell therapy.
Collapse
Affiliation(s)
- He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- Institute of Hematology, Zhejiang University, Hangzhou 310058, China
| | - Heng-wei Wu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- Institute of Hematology, Zhejiang University, Hangzhou 310058, China
| | - Yong-xian Hu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- Institute of Hematology, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
130
|
Yan L, Shang J, Shi X, Kang H, Liu W, Xu N, Liu Y, Chen G, Kang L, Gong F, Tang F, Yu L, Wu D, Fu C. Successful treatment of marrow failure after CARTs for myeloma by the infusion of cryopreserved stem cells. Am J Hematol 2020; 95:E20-E23. [PMID: 31637745 PMCID: PMC6972657 DOI: 10.1002/ajh.25664] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Lingzhi Yan
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of HematologyNational Clinical Research Center for Hematologic Diseases Suzhou China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University Suzhou China
| | - Jingjing Shang
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of HematologyNational Clinical Research Center for Hematologic Diseases Suzhou China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University Suzhou China
| | - Xiaolan Shi
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of HematologyNational Clinical Research Center for Hematologic Diseases Suzhou China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University Suzhou China
| | - Huizhu Kang
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of HematologyNational Clinical Research Center for Hematologic Diseases Suzhou China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University Suzhou China
| | - Wei Liu
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of HematologyNational Clinical Research Center for Hematologic Diseases Suzhou China
| | - Nan Xu
- Shanghai Unicar‐Therapy Bio‐medicine Technology Co., Ltd Shanghai China
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University Shanghai China
| | - Yong Liu
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of HematologyNational Clinical Research Center for Hematologic Diseases Suzhou China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University Suzhou China
| | - Guanghua Chen
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of HematologyNational Clinical Research Center for Hematologic Diseases Suzhou China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University Suzhou China
| | - Liqing Kang
- Shanghai Unicar‐Therapy Bio‐medicine Technology Co., Ltd Shanghai China
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University Shanghai China
| | - Feiran Gong
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of HematologyNational Clinical Research Center for Hematologic Diseases Suzhou China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University Suzhou China
| | - Fang Tang
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of HematologyNational Clinical Research Center for Hematologic Diseases Suzhou China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University Suzhou China
| | - Lei Yu
- Shanghai Unicar‐Therapy Bio‐medicine Technology Co., Ltd Shanghai China
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University Shanghai China
| | - Depei Wu
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of HematologyNational Clinical Research Center for Hematologic Diseases Suzhou China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University Suzhou China
| | - Chengcheng Fu
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of HematologyNational Clinical Research Center for Hematologic Diseases Suzhou China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University Suzhou China
| |
Collapse
|
131
|
Hosen N. Chimeric Antigen Receptor T-Cell Therapy for Multiple Myeloma. Cancers (Basel) 2019; 11:cancers11122024. [PMID: 31847470 PMCID: PMC6966463 DOI: 10.3390/cancers11122024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/27/2022] Open
Abstract
CD19 Chimeric antigen receptor (CAR) T cell therapy has been shown to be effective for B cell leukemia and lymphoma. Many researchers are now trying to develop CAR T cells for various types of cancer. For multiple myeloma (MM), B-cell maturation antigen (BCMA) has been recently proved to be a promising target. However, cure of MM is still difficult, and several other targets, for example immunoglobulin kappa chain, SLAM Family Member 7 (SLAMF7), or G-protein coupled receptor family C group 5 member D (GPRC5D), are being tested as targets for CAR T cells. We also reported that the activated integrin β7 can serve as a specific target for CAR T cells against MM, and are preparing a clinical trial. In this review, we summarized current status of CAR T cell therapy for MM and discussed about the future perspectives.
Collapse
Affiliation(s)
- Naoki Hosen
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Suita, 1-7 Yamada-Oka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
132
|
Immunotherapy for Multiple Myeloma. Cancers (Basel) 2019; 11:cancers11122009. [PMID: 31842518 PMCID: PMC6966649 DOI: 10.3390/cancers11122009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 01/08/2023] Open
Abstract
Despite therapeutic advances over the past decades, multiple myeloma (MM) remains a largely incurable disease with poor prognosis in high-risk patients, and thus new treatment strategies are needed to achieve treatment breakthroughs. MM represents various forms of impaired immune surveillance characterized by not only disrupted antibody production but also immune dysfunction of T, natural killer cells, and dendritic cells, although immunotherapeutic interventions such as allogeneic stem-cell transplantation and dendritic cell-based tumor vaccines were reported to prolong survival in limited populations of MM patients. Recently, epoch-making immunotherapies, i.e., immunomodulatory drug-intensified monoclonal antibodies, such as daratumumab combined with lenalidomide and chimeric antigen receptor T-cell therapy targeting B-cell maturation antigen, have been developed, and was shown to improve prognosis even in advanced-stage MM patients. Clinical trials using other antibody-based treatments, such as antibody drug-conjugate and bispecific antigen-directed CD3 T-cell engager targeting, are ongoing. The manipulation of anergic T-cells by checkpoint inhibitors, including an anti-T-cell immunoglobulin and ITIM domains (TIGIT) antibody, also has the potential to prolong survival times. Those new treatments or their combination will improve prognosis and possibly point toward a cure for MM.
Collapse
|
133
|
Sidana S, Shah N. CAR T-cell therapy: is it prime time in myeloma? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:260-265. [PMID: 31808895 PMCID: PMC6913444 DOI: 10.1182/hematology.2019000370] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Chimeric antigen receptor (CAR) T cells have shown promising activity in hematological malignancies and are being studied for the treatment of multiple myeloma, as well. B-cell maturation antigen, which is widely and almost exclusively expressed on plasma cells and B cells, is a promising target. Other targets being evaluated include CD19, CD38, CD138, signaling lymphocyte activation molecule or CS1, light chain, GPRC5D, and NKG2D. Early clinical studies have shown promising response rates in heavily pretreated patients, but relapses have occurred. Cytokine release syndrome and neurotoxicity have been observed in the majority of patients but are mostly grades 1 and 2. Relapse may be mediated by antigen escape and the limited persistence of CAR T cells. CAR T-cell constructs that target multiple antigens/epitopes or constructs with longer persistence due to a higher proportion of memory phenotype T cells may decrease the rates of relapse. Allogeneic CAR T cells that offer "off-the-shelf" options are also being developed. The challenges in integrating CAR T cells in myeloma therapy include disease relapse, adverse effects, cost, and identifying the right patient population. Longer-term data on efficacy and toxicity are needed before CAR T cells are ready for prime time in myeloma.
Collapse
Affiliation(s)
- Surbhi Sidana
- Department of Medicine, Stanford University, Stanford, CA; and
| | - Nina Shah
- Division of Hematology-Oncology, University of California, San Francisco, CA
| |
Collapse
|
134
|
Holstein SA, Lunning MA. CAR T‐Cell Therapy in Hematologic Malignancies: A Voyage in Progress. Clin Pharmacol Ther 2019; 107:112-122. [DOI: 10.1002/cpt.1674] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Sarah A. Holstein
- Division of Oncology and Hematology Department of Internal Medicine University of Nebraska Medical Center Omaha Nebraska USA
| | - Matthew A. Lunning
- Division of Oncology and Hematology Department of Internal Medicine University of Nebraska Medical Center Omaha Nebraska USA
| |
Collapse
|
135
|
Wu C, Zhang L, Brockman QR, Zhan F, Chen L. Chimeric antigen receptor T cell therapies for multiple myeloma. J Hematol Oncol 2019; 12:120. [PMID: 31752943 PMCID: PMC6873434 DOI: 10.1186/s13045-019-0823-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/07/2019] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy and remains incurable despite the advent of numerous new drugs such as proteasome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies. There is an unmet need to develop novel therapies for refractory/relapsed MM. In the past few years, chimeric antigen receptor (CAR)-modified T cell therapy for MM has shown promising efficacy in preclinical and clinical studies. Furthermore, the toxicities of CAR-T cell therapy are manageable. This article summarizes recent developments of CAR-T therapy in MM, focusing on promising targets, new technologies, and new research areas. Additionally, a comprehensive overview of antigen selection is presented along with preliminary results and future directions of CAR-T therapy development.
Collapse
Affiliation(s)
- Chao Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Lina Zhang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Qierra R Brockman
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation and Holden Comprehensive Cancer Center, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA.,Molecular Medicine Program, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA
| | - Fenghuang Zhan
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation and Holden Comprehensive Cancer Center, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA
| | - Lijuan Chen
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.
| |
Collapse
|
136
|
Sidana S, Shah N. CAR T-cell therapy: is it prime time in myeloma? Blood Adv 2019; 3:3473-3480. [PMID: 31714964 PMCID: PMC6855102 DOI: 10.1182/bloodadvances.2019000370] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/24/2019] [Indexed: 11/20/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells have shown promising activity in hematological malignancies and are being studied for the treatment of multiple myeloma, as well. B-cell maturation antigen, which is widely and almost exclusively expressed on plasma cells and B cells, is a promising target. Other targets being evaluated include CD19, CD38, CD138, signaling lymphocyte activation molecule or CS1, light chain, GPRC5D, and NKG2D. Early clinical studies have shown promising response rates in heavily pretreated patients, but relapses have occurred. Cytokine release syndrome and neurotoxicity have been observed in the majority of patients but are mostly grades 1 and 2. Relapse may be mediated by antigen escape and the limited persistence of CAR T cells. CAR T-cell constructs that target multiple antigens/epitopes or constructs with longer persistence due to a higher proportion of memory phenotype T cells may decrease the rates of relapse. Allogeneic CAR T cells that offer "off-the-shelf" options are also being developed. The challenges in integrating CAR T cells in myeloma therapy include disease relapse, adverse effects, cost, and identifying the right patient population. Longer-term data on efficacy and toxicity are needed before CAR T cells are ready for prime time in myeloma.
Collapse
Affiliation(s)
- Surbhi Sidana
- Department of Medicine, Stanford University, Stanford, CA; and
| | - Nina Shah
- Division of Hematology-Oncology, University of California, San Francisco, CA
| |
Collapse
|
137
|
Pont MJ, Hill T, Cole GO, Abbott JJ, Kelliher J, Salter AI, Hudecek M, Comstock ML, Rajan A, Patel BKR, Voutsinas JM, Wu Q, Liu L, Cowan AJ, Wood BL, Green DJ, Riddell SR. γ-Secretase inhibition increases efficacy of BCMA-specific chimeric antigen receptor T cells in multiple myeloma. Blood 2019; 134:1585-1597. [PMID: 31558469 PMCID: PMC6871311 DOI: 10.1182/blood.2019000050] [Citation(s) in RCA: 214] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 08/20/2019] [Indexed: 12/15/2022] Open
Abstract
B-cell maturation antigen (BCMA) is a validated target for chimeric antigen receptor (CAR) T-cell therapy in multiple myeloma (MM). Despite promising objective response rates, most patients relapse, and low levels of BCMA on a subset of tumor cells has been suggested as a probable escape mechanism. BCMA is actively cleaved from the tumor cell surface by the ubiquitous multisubunit γ-secretase (GS) complex, which reduces ligand density on tumor cells for CAR T-cell recognition and releases a soluble BCMA (sBCMA) fragment capable of inhibiting CAR T-cell function. Sufficient sBCMA can accumulate in the bone marrow of MM patients to inhibit CAR T-cell recognition of tumor cells, and potentially limit efficacy of BCMA-directed adoptive T-cell therapy. We investigated whether blocking BCMA cleavage by small-molecule GS inhibitors (GSIs) could augment BCMA-targeted CAR T-cell therapy. We found that exposure of myeloma cell lines and patient tumor samples to GSIs markedly increased surface BCMA levels in a dose-dependent fashion, concurrently decreased sBCMA concentrations, and improved tumor recognition by CAR T cells in vitro. GSI treatment of MM tumor-bearing NOD/SCID/γc-/- mice increased BCMA expression on tumor cells, decreased sBCMA in peripheral blood, and improved antitumor efficacy of BCMA-targeted CAR T-cell therapy. Importantly, short-term GSI administration to MM patients markedly increases the percentage of BCMA+ tumor cells, and the levels of BCMA surface expression in vivo. Based on these data, a US Food and Drug Administration (FDA)-approved clinical trial has been initiated, combining GSI with concurrent BCMA CAR T-cell therapy. This trial was registered at www.clinicaltrials.gov as #NCT03502577.
Collapse
Affiliation(s)
- Margot J Pont
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Tyler Hill
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Gabriel O Cole
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Joe J Abbott
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jessica Kelliher
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Alexander I Salter
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany; and
| | - Melissa L Comstock
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Anusha Rajan
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Jenna M Voutsinas
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Qian Wu
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Lingfeng Liu
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Andrew J Cowan
- Department of Medicine, University of Washington, Seattle, WA
| | - Brent L Wood
- Department of Medicine, University of Washington, Seattle, WA
| | - Damian J Green
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Stanley R Riddell
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
138
|
Lin Q, Zhao J, Song Y, Liu D. Recent updates on CAR T clinical trials for multiple myeloma. Mol Cancer 2019; 18:154. [PMID: 31684964 PMCID: PMC6829852 DOI: 10.1186/s12943-019-1092-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 10/18/2019] [Indexed: 12/22/2022] Open
Abstract
Proteasome inhibitors, immunomodulatory agents and monoclonal antibodies have dramatically changed the natural history of multiple myeloma (MM). However, most patients eventually suffer a relapse and succumb to the disease. Chimeric antigen receptor (CAR) engineered T cells targeting B cell maturation antigen (BCMA), CD138, CS1 glycoprotein antigen (SLAMF7) and light chains are in active development for therapy of refractory /relapsed (RR) MM. CD19- targeted CAR T cells in conjunction with autologous stem cell transplantation also showed activity in RRMM. Dual- target CAR T cells are in clinical trials for RRMM. This review summarized the recent updates of ongoing CAR T clinical trials for multiple myeloma.
Collapse
Affiliation(s)
- Quande Lin
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| | - Juanjuan Zhao
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| | - Yongping Song
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| | - Delong Liu
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| |
Collapse
|
139
|
Feinberg D, Paul B, Kang Y. The promise of chimeric antigen receptor (CAR) T cell therapy in multiple myeloma. Cell Immunol 2019; 345:103964. [PMID: 31492448 PMCID: PMC6832886 DOI: 10.1016/j.cellimm.2019.103964] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/08/2019] [Accepted: 08/10/2019] [Indexed: 12/19/2022]
Abstract
A cure for multiple myeloma (MM), a malignancy of plasma cells, remains elusive. Nearly all myeloma patients will eventually relapse and develop resistance to currently available treatments. There is an unmet medical need to develop novel and effective therapies that can induce sustained responses. Early phase clinical trials using chimeric antigen receptor (CAR) T cell therapy have shown great promise in the treatment of relapsed and/or refractory MM. In this review article, we provide an overview of the CAR constructs, the gene transfer vector systems, and approaches for T cell activation and expansion. We then summarize the outcomes of several early phase clinical trials of CAR T cell therapy in MM and the novel CAR T targets that are under development. Finally, we explore the potential mechanisms that result in disease relapse after CAR T therapy and propose future directions in CAR T therapy in MM.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Hematopoietic Stem Cell Transplantation/methods
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Multiple Myeloma/immunology
- Multiple Myeloma/metabolism
- Multiple Myeloma/therapy
- Neoplasm Recurrence, Local
- Outcome Assessment, Health Care
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Daniel Feinberg
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| | - Barry Paul
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
140
|
Wang H, Lin P. Flow Cytometric Immunophenotypic Analysis in the Diagnosis and Prognostication of Plasma Cell Neoplasms. CYTOMETRY PART B-CLINICAL CYTOMETRY 2019; 96:338-350. [DOI: 10.1002/cyto.b.21844] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Hao‐Wei Wang
- Flow Cytometry and Hematopathology Section, Laboratory of PathologyCCR, NCI, NIH Bethesda Maryland 20892
| | - Pei Lin
- Department of HematopathologyThe University of Texas MD Anderson Cancer Center Houston Texas 77030
| |
Collapse
|
141
|
Timmers M, Roex G, Wang Y, Campillo-Davo D, Van Tendeloo VFI, Chu Y, Berneman ZN, Luo F, Van Acker HH, Anguille S. Chimeric Antigen Receptor-Modified T Cell Therapy in Multiple Myeloma: Beyond B Cell Maturation Antigen. Front Immunol 2019; 10:1613. [PMID: 31379824 PMCID: PMC6646459 DOI: 10.3389/fimmu.2019.01613] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 06/28/2019] [Indexed: 12/27/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T cell therapy is a rapidly emerging immunotherapeutic approach that is revolutionizing cancer treatment. The impressive clinical results obtained with CAR-T cell therapy in patients with acute lymphoblastic leukemia and lymphoma have fueled the development of CAR-T cells targeting other malignancies, including multiple myeloma (MM). The field of CAR-T cell therapy for MM is still in its infancy, but remains promising. To date, most studies have been performed with B cell maturation antigen (BCMA)-targeted CARs, for which high response rates have been obtained in early-phase clinical trials. However, responses are usually temporary, and relapses have frequently been observed. One of the major reasons for relapse is the loss or downregulation of BCMA expression following CAR-T therapy. This has fostered a search for alternative target antigens that are expressed on the MM cell surface. In this review, we provide an overview of myeloma target antigens other than BCMA that are currently being evaluated in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Marijke Timmers
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Gils Roex
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Yuedi Wang
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Viggo F I Van Tendeloo
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Yiwei Chu
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Zwi N Berneman
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Feifei Luo
- Biotherapy Research Center, Fudan University, Shanghai, China.,Department of Digestive Diseases, Huashan Hospital of Fudan University, Shanghai, China
| | - Heleen H Van Acker
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Sébastien Anguille
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
142
|
Abstract
Introduction: Immunotherapy has revolutionized the treatment of cancer. Antibodies, antibody drug conjugates, and bispecific antibodies have improved outcomes in various cancers especially lymphomas. Chimeric antigen receptor T cell (CAR-T) is a step forward in the immunotherapy paradigm for the treatment of Lymphomas. Recently, two CAR-T products, Tisagenlecleucel and Axicabtagene ciloleucel, were approved by the US FDA. While it is exciting to have such novel treatment available, the challenges of production, administration, related toxicity, and cost remain. Specific toxicities related to CAR-T like Cytokine Release Syndrome (CRS) and Immune Effector Cell-Associated Neurotoxicity Syndrome (ICANS) could be fatal and need close monitoring and prompt treatment to avoid mortality and improve efficacy of the treatment. Areas covered: In this article, the authors discuss receptor constructs, administration, toxicities, efficacy and reimbursement of CAR-T treatment. Expert opinion: Since approval of CAR-T treatment, cost of therapy and reimbursement have been a big challenge in implementation of CAR-T. This has triggered cost-effective analysis and nationwide discussions about the reimbursement process of such treatment. In spite of these challenges, CAR-T treatment is a huge step forward with a very bright future. Novel CAR-T targeting a variety of antigens in different cancers seems promising in near future.
Collapse
Affiliation(s)
- Jennifer Kelly Anderson
- a Hematology and Oncology fellow, Department of Hematology and Oncology , University of Alabama , Birmingham , AL , USA
| | - Amitkumar Mehta
- b Department of Hematology and Oncology , University of Alabama , Birmingham , AL , USA
| |
Collapse
|
143
|
Elmaagacli AH, Salwender H, Jehn C, Dahmash F, Singh A, Wilson O, Pannenbeckers M, Niggemann C, Vierbuchen M. Strong expression of SLAMF7 in natural killer/T-cell lymphoma and large granular lymphocyte leukemia - a prominent biomarker and potential target for anti-SLAMF7 antibody therapy. Leuk Lymphoma 2019; 60:3335-3338. [PMID: 31164030 DOI: 10.1080/10428194.2019.1623887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Ahmet H Elmaagacli
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Hans Salwender
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Christian Jehn
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Farouk Dahmash
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Anju Singh
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Oliver Wilson
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Marc Pannenbeckers
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | | | | |
Collapse
|
144
|
Giuliani N, Accardi F, Marchica V, Dalla Palma B, Storti P, Toscani D, Vicario E, Malavasi F. Novel targets for the treatment of relapsing multiple myeloma. Expert Rev Hematol 2019; 12:481-496. [PMID: 31125526 DOI: 10.1080/17474086.2019.1624158] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Introduction: Multiple myeloma (MM) is characterized by the high tendency to relapse and develop drug resistance. Areas covered: This review focused on the main novel targets identified to design drugs for the treatment of relapsing MM patients. CD38 and SLAMF7 are the main surface molecules leading to the development of monoclonal antibodies (mAbs) recently approved for the treatment of relapsing MM patients. B cell maturation antigen (BCMA) is a suitable target for antibody-drug conjugates, bispecific T cell engager mAbs and Chimeric Antigen Receptor (CAR)-T cells. Moreover, the programmed cell death protein 1 (PD)-1/PD-Ligand (PD-L1) expression profile by MM cells and their microenvironment and the use of immune checkpoints inhibitors in MM patients are reported. Finally, the role of histone deacetylase (HDAC), B cell lymphoma (BCL)-2 family proteins and the nuclear transport protein exportin 1 (XPO1) as novel targets are also underlined. The clinical results of the new inhibitors in relapsing MM patients are discussed. Expert opinion: CD38, SLAMF7, and BCMA are the main targets for different immunotherapeutic approaches. Selective inhibitors of HDAC6, BCL-2, and XPO1 are new promising compounds under clinical investigation in relapsing MM patients.
Collapse
Affiliation(s)
- Nicola Giuliani
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Fabrizio Accardi
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Valentina Marchica
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | | | - Paola Storti
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Denise Toscani
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Emanuela Vicario
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Fabio Malavasi
- b Department of Medical Science , University of Turin , Turin , Italy
| |
Collapse
|
145
|
Khan AM, Devarakonda S, Bumma N, Chaudhry M, Benson DM. Potential of NK cells in multiple Myeloma therapy. Expert Rev Hematol 2019; 12:425-435. [PMID: 31070067 DOI: 10.1080/17474086.2019.1617128] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Despite rapid advances in myeloma treatment with the development of new drugs, curative therapies remain elusive. Relapsed/refractory disease related to progressive dysregulation of immune system and acquired genetic abnormalities continues to be a major obstacle in achieving cure. Immune-based therapy harnessing the host defense mechanism of natural killer (NK) cells is a promising avenue in the treatment of myeloma. Areas covered: Here, we discuss the biology and cytotoxic activity of NK cells and the potential role of these innate immune cells in defense against cancer and specifically multiple myeloma. We also discuss the role of NK cells in the anti-myeloma effects of autologous and allogeneic stem cell transplantation, various novel drugs, and treatment modalities such as chimeric antigen receptor therapy. Immune evasion, either directly or indirectly involving NK cell dysfunction, may be a key and under-recognized mechanism in myeloma progression. We reviewed extensive literature identified using the keywords immunotherapy, natural killer cells, and multiple myeloma. Expert opinion: Novel treatment approaches in myeloma utilizing the immunomodulatory and cytotoxic properties of NK cells to eradicate resistant and quiescent clones could pave the way for potentially curative interventions.
Collapse
Affiliation(s)
- Abdullah M Khan
- a Division of Hematology, Department of Medicine , The Ohio State University Comprehensive Cancer Center , Columbus , OH , USA
| | - Srinivas Devarakonda
- a Division of Hematology, Department of Medicine , The Ohio State University Comprehensive Cancer Center , Columbus , OH , USA
| | - Naresh Bumma
- a Division of Hematology, Department of Medicine , The Ohio State University Comprehensive Cancer Center , Columbus , OH , USA
| | - Maria Chaudhry
- a Division of Hematology, Department of Medicine , The Ohio State University Comprehensive Cancer Center , Columbus , OH , USA
| | - Don M Benson
- a Division of Hematology, Department of Medicine , The Ohio State University Comprehensive Cancer Center , Columbus , OH , USA
| |
Collapse
|
146
|
Multiple Myeloma, Targeting B-Cell Maturation Antigen With Chimeric Antigen Receptor T-Cells. Cancer J 2019; 25:208-216. [DOI: 10.1097/ppo.0000000000000379] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
147
|
Hagen PA, Stiff P. The Role of Salvage Second Autologous Hematopoietic Cell Transplantation in Relapsed Multiple Myeloma. Biol Blood Marrow Transplant 2019; 25:e98-e107. [DOI: 10.1016/j.bbmt.2018.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 12/05/2018] [Indexed: 10/27/2022]
|
148
|
Kloess S, Kretschmer A, Stahl L, Fricke S, Koehl U. CAR-Expressing Natural Killer Cells for Cancer Retargeting. Transfus Med Hemother 2019; 46:4-13. [PMID: 31244577 DOI: 10.1159/000495771] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/23/2018] [Indexed: 12/15/2022] Open
Abstract
Since the approval in 2017 and the outstanding success of Kymriah® and Yescarta®, the number of clinical trials investigating the safety and efficacy of chimeric antigen receptor-modified autologous T cells has been constantly rising. Currently, more than 200 clinical trials are listed on clinicaltrial.gov. In contrast to CAR-T cells, natural killer (NK) cells can be used from allogeneic donors as an "off the shelf product" and provide alternative candidates for cancer retargeting. This review summarises preclinical results of CAR-engineered NK cells using both primary human NK cells and the cell line NK-92, and provides an overview about the first clinical CAR-NK cell studies targeting haematological malignancies and solid tumours, respectively.
Collapse
Affiliation(s)
- Stephan Kloess
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany.,Institute for Cellular Therapeutics, ATMP-GMPDU, Hannover Medical School, Hannover, Germany
| | - Anna Kretschmer
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Lilly Stahl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Ulrike Koehl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany.,Institute of Clinical Immunology, Faculty of Medicine, University Leipzig, Leipzig, Germany.,Institute for Cellular Therapeutics, ATMP-GMPDU, Hannover Medical School, Hannover, Germany
| |
Collapse
|
149
|
[CAR-T cells: Lymphocytes that express a chimeric antigen receptor]. Rev Med Interne 2019; 40:545-552. [PMID: 30686549 DOI: 10.1016/j.revmed.2018.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/30/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
CAR-T cells are genetically modified human lymphocytes and gene therapy medicinal products. They are developed to treat cancers that express a membrane antigen targeted by the CAR. The FDA approved the two first-in-class medicinal products in 2017 and EMA in August 2018; both are autologous CAR-T cells targeting CD19 that is expressed at the surface of normal B-cells throughout their differentiation, and on B-cell lymphoid malignancies. Clinical efficacy was demonstrated for B-cell acute lymphoblastic leukemias, non-Hodgkin's lymphoma and chronic lymphocytic leukemia, although the marketing authorizations are less liberal in terms of indications. Manufacturing of these personalized treatments necessitates that a novel organization and supply chain be set in place, to ensure product preservation, patient safety and compliance with complex regulatory requirements. Side effects are commensurate with clinical efficacy and can be life-threatening: proper management imposes tight coordination between various specialists, particularly between hematologists and intensive care practitioners. High pricing for these treatments is part of a long-term trend for increasing costs of innovations in hematology and oncology; it questions the ability of healthcare systems to sustain their reimbursement.
Collapse
|
150
|
Franssen LE, Mutis T, Lokhorst HM, van de Donk NWCJ. Immunotherapy in myeloma: how far have we come? Ther Adv Hematol 2019; 10:2040620718822660. [PMID: 30719268 PMCID: PMC6348514 DOI: 10.1177/2040620718822660] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/06/2018] [Indexed: 12/31/2022] Open
Abstract
The treatment of multiple myeloma (MM) has evolved substantially over the past decades, leading to a significantly improved outcome of MM patients. The introduction of high-dose therapy, especially, and autologous stem cell transplantation, as well as the development of new drugs, such as immunomodulatory drugs (IMiDs) and proteasome inhibitors have contributed to the improvement in survival. However, eventually most MM patients relapse, which indicates that there is a need for new agents and novel treatment strategies. Importantly, the long-term survival in a subset of MM patients after allogeneic stem cell transplantation illustrates the potential of immunotherapy in MM, but allogeneic stem cell transplantation is also associated with a high rate of treatment-related mortality. Recently, a better insight into several immune-evasion mechanisms, which contribute to tumor progression, has resulted in the development of active and well-tolerated novel forms of immunotherapy. These immunotherapeutic agents can be used as monotherapy, or, even more successfully, in combination with other established anti-MM agents to further improve depth and duration of response by preventing the outgrowth of resistant clones. This review will discuss the mechanisms used by MM cells to evade the immune system, and also provide an overview of currently approved immunotherapeutic drugs, such as IMiDs (e.g. lenalidomide and pomalidomide) and monoclonal antibodies that target cell surface antigens present on the MM cell (e.g. elotuzumab and daratumumab), as well as novel immunotherapies (e.g. chimeric antigen receptor T-cells, bispecific antibodies and checkpoint inhibitors) currently in clinical development in MM.
Collapse
Affiliation(s)
- Laurens E Franssen
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Tuna Mutis
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Henk M Lokhorst
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|