1
|
Deng L, Xu G. Update on the Application of Monoclonal Antibody Therapy in Primary Membranous Nephropathy. Drugs 2023; 83:507-530. [PMID: 37017915 DOI: 10.1007/s40265-023-01855-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
When first introduced, rituximab (RTX), a chimeric anti-CD20 monoclonal antibody, brought about an alternative therapeutic paradigm for primary membranous nephropathy (PMN). Rituximab was shown to be effective and safe in PMN patients with kidney dysfunction, with. patients receiving second-line rituximab therapy achieving remission as effectively as those patients who had not previously received immunotherapy. No safety issues were reported. The B cell-driven protocol seems to be as efficient as the 375 mg/m2 × 4 regimen or 1 g × 2 regimen in achieving B cell depletion and remission, but patients with high M-type phospholipase A2 receptor (PLA2R) antibody levels may benefit from a higher dose of rituximab. While rituximab added another therapeutic option to the treatment regimen, it does have limitations as 20 to 40% of patients do not respond. Not all patients respond to RTX therapy for lymphoproliferative disorders either, therefore further novel anti-CD20 monoclonal antibodies have been developed and these may provide alternative therapeutic options for PMN. Ofatumumab, a fully human monoclonal antibody, specifically recognizes an epitope encompassing both the small and large extracellular loops of the CD20 molecule, resulting in increased complement-dependent cytotoxic activity. Ocrelizumab binds an alternative but overlapping epitope region to rituximab and displays enhanced antibody-dependent cellular cytotoxic (ADCC) activities. Obinutuzumab is designed to have a modified elbow-hinge amino acid sequence, leading to increased direct cell death induction and ADCC activities. In PMN clinical studies, ocrelizumab and obinutuzumab showed promising results, while ofatumumab displayed mixed results. However, there is a lack of randomized controlled trials with large samples, especially direct head-to-head comparisons. Alternative molecular mechanisms have been suggested in this context to explore novel therapeutic strategies. B cell activator-targeted, plasma cell-targeted and complement-directed treatments may lead to novel therapy paradigms for PMN. Exploratory strategies for the use of drugs with different mechanisms, such as a combination of rituximab and cyclophosphamide and a steroid, a combination of rituximab and a calcineurin inhibitor, may provide more rapid and efficient remission, but the combination of standard immunosuppression with rituximab could increase infection risk.
Collapse
Affiliation(s)
- Le Deng
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
2
|
Dupilumab-Induced, Tralokinumab-Induced, and Belantamab Mafodotin-Induced Adverse Ocular Events-Incidence, Etiology, and Management. Cornea 2023; 42:507-519. [PMID: 36525340 PMCID: PMC9973444 DOI: 10.1097/ico.0000000000003162] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/15/2022] [Indexed: 12/23/2022]
Abstract
ABSTRACT Emerging monoclonal antibody therapies are assuming greater importance in the management of severe and refractory forms of immunity-driven and oncological disorders. However, some have been found to induce adverse ocular events (AOEs) leading to discontinuation of treatment or additional multidisciplinary management. We present the current knowledge concerning AOEs associated with 3 monoclonal antibody therapies: dupilumab, tralokinumab, and belantamab mafodotin. We examine the manifestations of their AOEs, proposed pathophysiological mechanisms, and current treatment recommendations. We identified and reviewed all studies for dupilumab, tralokinumab, and belantamab mafodotin using the keywords "dupilumab," "tralokinumab," "belantamab mafodotin," "conjunctivitis," and "keratopathy" from January 2016 to November 2021. Conjunctivitis was the most frequently reported AOE in patients with atopic dermatitis receiving dupilumab or tralokinumab. Mild cases were managed with warm compresses for associated meibomian gland dysfunction, artificial tears, and antihistamine/mast cell stabilizer eye drops. In more severe cases, additional anti-inflammatory therapy, with corticosteroid eye drops or ointments, or topical calcineurin inhibitors-such as tacrolimus or ciclosporin-were required. Patients with resistant or refractory multiple myeloma treated with belantamab mafodotin often developed keratopathy, which could necessitate contact lens fitting, or for cycles of belantamab mafodotin to be delayed.
Collapse
|
3
|
Sioud M, Olberg A. Antibody Surface Profiling Identifies Glycoforms in Multiple Myeloma as Targets for Immunotherapy: From Antibody Derivatives to Mimetic Peptides for Killing Tumor Cells. Cancers (Basel) 2023; 15:cancers15071934. [PMID: 37046595 PMCID: PMC10093763 DOI: 10.3390/cancers15071934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/15/2023] [Accepted: 03/18/2023] [Indexed: 04/14/2023] Open
Abstract
Despite therapeutic advances in recent years, there are still unmet medical needs for patients with multiple myeloma (MM). Hence, new therapeutic strategies are needed. Using phage display for screening a large repertoire of single chain variable fragments (scFvs), we isolated several candidates that recognize a heavily sulfated MM-specific glycoform of the surface antigen syndecan-1 (CD138). One of the engineered scFv-Fc antibodies, named MM1, activated NK cells and induced antibody-dependent cellular cytotoxicity against MM cells. Analysis of the binding specificity by competitive binding assays with various glycan ligands identified N-sulfation of glucosamine units as essential for binding. Additionally, site-directed mutagenesis revealed that the amino acids arginine and histidine in the complementarily determining regions (CDRs) 2 and 3 of the heavy chain are important for binding. Based on this observation, a heavy-chain antibody, known as a nanobody, and a peptide mimicking the CDR loop sequences were designed. Both variants exhibited high affinity and specificity to MM cells as compared to blood lymphocytes. Specific killing of MM cells was achieved by conjugating the CDR2/3 mimic peptide to a pro-apoptotic peptide (KLAKLAK)2. In a co-culture model, the fusion peptide killed MM cells, while leaving normal peripheral blood mononuclear cells unaffected. Collectively, the development of antibodies and peptides that detect tumor-specific glycoforms of therapeutic targets holds promise for improving targeted therapies and tumor imaging.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Cancer Immunology, Division of Cancer Medicine, Oslo University Hospital-Radiumhospitalet, Ullernchausseen 70, 0379 Oslo, Norway
| | - Anniken Olberg
- Department of Cancer Immunology, Division of Cancer Medicine, Oslo University Hospital-Radiumhospitalet, Ullernchausseen 70, 0379 Oslo, Norway
| |
Collapse
|
4
|
Seervai RNH, Friske SK, Chu EY, Phillips R, Nelson KC, Huen A, Cho WC, Aung PP, Torres-Cabala CA, Prieto VG, Curry JL. The diverse landscape of dermatologic toxicities of non-immune checkpoint inhibitor monoclonal antibody-based cancer therapy. J Cutan Pathol 2023; 50:72-95. [PMID: 36069496 DOI: 10.1111/cup.14327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/30/2022] [Accepted: 09/04/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND Since their first approval 25 years ago, monoclonal antibodies (mAbs) have become important targeted cancer therapeutics. However, dermatologic toxicities associated with non-immune checkpoint inhibitor (non-ICI) mAbs may complicate the course of cancer treatment. Data on the incidence and types of these reactions are limited. METHODS A comprehensive review was conducted on dermatologic toxicities associated with different classes of non-ICI mAbs approved for treatment of solid tumors and hematologic malignancies. The review included prospective Phase 1, 2, and 3 clinical trials; retrospective literature reviews; systematic reviews/meta-analyses; and case series/reports. RESULTS Dermatologic toxicities were associated with several types of non-ICI mAbs. Inflammatory reactions were the most common dermatologic toxicities, manifesting as maculopapular, urticarial, papulopustular/acneiform, and lichenoid/interface cutaneous adverse events (cAEs) with non-ICI mAbs. Immunobullous reactions were rare and a subset of non-ICI mAbs were associated with the development of vitiligo cAEs. CONCLUSION Dermatologic toxicities of non-ICI mAbs are diverse and mostly limited to inflammatory reactions. Awareness of the spectrum of the histopathologic patterns of cAE from non-ICI mAbs therapy is critical in the era of oncodermatology and oncodermatopathology.
Collapse
Affiliation(s)
- Riyad N H Seervai
- Internal Medicine Residency Program, Providence Portland Medical Center, Portland, Oregon, USA.,Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, USA.,Department of Dermatology, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah K Friske
- Department of Dermatology, Baylor College of Medicine, Houston, Texas, USA
| | - Emily Y Chu
- Department of Dermatology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rhea Phillips
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kelly C Nelson
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Auris Huen
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Woo Cheal Cho
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Phyu P Aung
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Carlos A Torres-Cabala
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Victor G Prieto
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jonathan L Curry
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
5
|
Ghosh K, Ghosh K. Monoclonal antibodies used for the management of haematological disorders. Expert Rev Hematol 2022; 15:443-455. [PMID: 35504000 DOI: 10.1080/17474086.2022.2073213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Monoclonal antibodies Ab (MoAb) are increasingly becoming part of therapeutic armamentarium for haematologists and haemato-oncologists. This review brings together commonly used antibodies in one place for brevity and novel understanding. AREAS COVERED Pubmed and Scopus databases were explored focusing on MoAb in clinical haematological practice. Emphasis was given to current review articles. The data base was searched from 1997 till present. 24 different antibodies, most of which are in use were discussed. Antibodies are used for diverse conditions i.e. malignant and benign haematological conditions, treatment at various phases of stem cell transplantation. These antibodies were used both alone or in combination with various chemotherapy, targeted small molecules or as immunoconjugates. Some of the side effect profiles of these antibodies were common and some were unique. Unusual infections or organ dysfunctions were noted. Improved function of antibodies by protein engineering is also advancing rapidly. Dosage, frequency and route of administration depended on the convenience and condition for which the antibody is used. EXPERT OPINION : MoAbs are increasingly used in haematology practice either alone or in combination with other types of therapy for improved out come in various haematological conditions.
Collapse
Affiliation(s)
- Kanjaksha Ghosh
- National Institute of Immunohaematology. 13th fl. KEM Hospital MS Building, Parel, Mumbai 400012. India
| | - Kinjalka Ghosh
- Department of Clinical Biochemistry , Tata Memorial Hospital. & Homi Bhaba National Institute. Parel, Mumbai 400012.India
| |
Collapse
|
6
|
Teicher BA, Morris J. Antibody-Drug Conjugate Targets, Drugs and Linkers. Curr Cancer Drug Targets 2022; 22:463-529. [PMID: 35209819 DOI: 10.2174/1568009622666220224110538] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/22/2021] [Accepted: 11/09/2021] [Indexed: 11/22/2022]
Abstract
Antibody-drug conjugates offer the possibility of directing powerful cytotoxic agents to a malignant tumor while sparing normal tissue. The challenge is to select an antibody target expressed exclusively or at highly elevated levels on the surface of tumor cells and either not all or at low levels on normal cells. The current review explores 78 targets that have been explored as antibody-drug conjugate targets. Some of these targets have been abandoned, 9 or more are the targets of FDA-approved drugs, and most remain active clinical interest. Antibody-drug conjugates require potent cytotoxic drug payloads, several of these small molecules are discussed, as are the linkers between the protein component and small molecule components of the conjugates. Finally, conclusions regarding the elements for the successful antibody-drug conjugate are discussed.
Collapse
Affiliation(s)
- Beverly A Teicher
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| | - Joel Morris
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| |
Collapse
|
7
|
Girmenia C, Cavo M, Corso A, Di Raimondo F, Musto P, Offidani M, Petrucci MT, Rosato A, Barosi G. Management of infectious risk of daratumumab therapy in multiple myeloma: a consensus-based position paper from an ad hoc Italian expert panel. Crit Rev Oncol Hematol 2022; 172:103623. [PMID: 35151867 DOI: 10.1016/j.critrevonc.2022.103623] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/27/2022] [Accepted: 02/07/2022] [Indexed: 12/28/2022] Open
Abstract
Daratumumab is approved for newly diagnosed or relapsed/refractory multiple myeloma (MM). The use of daratumumab has improved patient outcomes but has changed the frequency and epidemiology of infections. However, the optimal approach to prophylaxis and supportive therapy for daratumumab-emergent infections is unknown and represents an unmet clinical need in MM. Consequently, an expert panel convened to compose recommendations for optimal infection control in patient candidates to or under daratumumab treatment for MM. Scientific evidence on infections secondary to daratumumab was evaluated, and a consensus was developed by group discussion for key questions selected according to the clinical relevance. The following key issues were addressed: infectious risk assessment and risk stratification, infection mitigation strategies, and management of infectious complications in patients with MM treated with daratumumab.
Collapse
Affiliation(s)
- Corrado Girmenia
- Dipartimento di Ematologia, Oncologia, e Dermatologia, AOU Policlinico Umberto I, Sapienza Università di Roma, Rome, Italy.
| | - Michele Cavo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli",Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | | | - Francesco Di Raimondo
- Dipartimento di specialità medico-chirurgiche - sezione di Ematologia, Università di Catania. Divisione di Ematologia con Trapianto di midollo osseo - Azienda ospedaliero, universitaria Policlinico Vittorio Emanuele, Catania, Italy
| | - Pellegrino Musto
- Department of Emergency and Organ Transplantation, "Aldo Moro" University School of Medicine, Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico, Bari, Italy
| | - Massimo Offidani
- Clinica di Ematologia, AOU Ospedali Riuniti di Ancona, Ancona, Italy
| | - Maria Teresa Petrucci
- Dipartimento di Ematologia, Oncologia, e Dermatologia, AOU Policlinico Umberto I, Sapienza Università di Roma, Rome, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua. Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis. IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| |
Collapse
|
8
|
Abramson HN. Immunotherapy of Multiple Myeloma: Promise and Challenges. Immunotargets Ther 2021; 10:343-371. [PMID: 34527606 PMCID: PMC8437262 DOI: 10.2147/itt.s306103] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022] Open
Abstract
Whereas the treatment of MM was dependent solely on alkylating agents and corticosteroids during the prior three decades, the landscape of therapeutic measures to treat the disease began to expand enormously early in the current century. The introduction of new classes of small-molecule drugs, such as proteasome blockers (bortezomib and carfilzomib), immunomodulators (lenalidomide and pomalidomide), nuclear export inhibitors (selinexor), and histone deacetylase blockers (panobinostat), as well as the application of autologous stem cell transplantation (ASCT), resulted in a seismic shift in how the disease is treated. The picture changed dramatically once again starting with the 2015 FDA approval of two monoclonal antibodies (mAbs) - the anti-CD38 daratumumab and the anti-SLAMF7 elotuzumab. Daratumumab, in particular, has had a great impact on MM therapy and today is often included in various regimens to treat the disease, both in newly diagnosed cases and in the relapse/refractory setting. Recently, other immunotherapies have been added to the arsenal of drugs available to fight this malignancy. These include isatuximab (also anti-CD38) and, in the past year, the antibody-drug conjugate (ADC) belantamab mafodotin and the chimeric antigen receptor (CAR) T-cell product idecabtagene vicleucel (ide-cel). While the accumulated benefits of these newer agents have resulted in a doubling of the disease's five-year survival rate to more than 5 years and improved quality of life, the disease remains incurable. Almost without exception patients experience relapse and/or become refractory to the drugs used, making the search for innovative therapies all the more essential. This review covers the current scope of anti-myeloma immunotherapeutic agents, both those in clinical use and on the horizon, including naked mAbs, ADCs, bi- and multi-targeted mAbs, and CAR T-cells. Emphasis is placed on the benefits of each along with the challenges that need to be overcome if MM is to be considered curable in the future.
Collapse
Affiliation(s)
- Hanley N Abramson
- Wayne State University, Department of Pharmaceutical Sciences, Detroit, MI, 48201, USA
| |
Collapse
|
9
|
Romano A, Storti P, Marchica V, Scandura G, Notarfranchi L, Craviotto L, Di Raimondo F, Giuliani N. Mechanisms of Action of the New Antibodies in Use in Multiple Myeloma. Front Oncol 2021; 11:684561. [PMID: 34307150 PMCID: PMC8297441 DOI: 10.3389/fonc.2021.684561] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/09/2021] [Indexed: 12/19/2022] Open
Abstract
Monoclonal antibodies (mAbs) directed against antigen-specific of multiple myeloma (MM) cells have Fc-dependent immune effector mechanisms, such as complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP), but the choice of the antigen is crucial for the development of effective immuno-therapy in MM. Recently new immunotherapeutic options in MM patients have been developed against different myeloma-related antigens as drug conjugate-antibody, bispecific T-cell engagers (BiTEs) and chimeric antigen receptor (CAR)-T cells. In this review, we will highlight the mechanism of action of immuno-therapy currently available in clinical practice to target CD38, SLAMF7, and BCMA, focusing on the biological role of the targets and on mechanisms of actions of the different immunotherapeutic approaches underlying their advantages and disadvantages with critical review of the literature data.
Collapse
Affiliation(s)
- Alessandra Romano
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| | - Paola Storti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Grazia Scandura
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| | | | - Luisa Craviotto
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Francesco Di Raimondo
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
- U.O.C. Ematologia, A.O.U. Policlinico–San Marco, Catania, Italy
| | | |
Collapse
|
10
|
Nasarre P, Garcia DI, Siegel JB, Bonilla IV, Mukherjee R, Hilliard E, Chakraborty P, Nasarre C, Yustein JT, Lang M, Jaffa AA, Mehrotra S, Klauber-DeMore N. Overcoming PD-1 Inhibitor Resistance with a Monoclonal Antibody to Secreted Frizzled-Related Protein 2 in Metastatic Osteosarcoma. Cancers (Basel) 2021; 13:cancers13112696. [PMID: 34070758 PMCID: PMC8199140 DOI: 10.3390/cancers13112696] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 12/20/2022] Open
Abstract
Secreted frizzled-related protein 2 (SFRP2) promotes the migration/invasion of metastatic osteosarcoma (OS) cells and tube formation by endothelial cells. However, its function on T-cells is unknown. We hypothesized that blocking SFRP2 with a humanized monoclonal antibody (hSFRP2 mAb) can restore immunity by reducing CD38 and PD-1 levels, ultimately overcoming resistance to PD-1 inhibitors. Treating two metastatic murine OS cell lines in vivo, RF420 and RF577, with hSFRP2 mAb alone led to a significant reduction in the number of lung metastases, compared to IgG1 control treatment. While PD-1 mAb alone had minimal effect, hSFRP2 mAb combination with PD-1 mAb had an additive antimetastatic effect. This effect was accompanied by lower SFRP2 levels in serum, lower CD38 levels in tumor-infiltrating lymphocytes and T-cells, and lower PD-1 levels in T-cells. In vitro data confirmed that SFRP2 promotes NFATc3, CD38 and PD-1 expression in T-cells, while hSFRP2 mAb treatment counteracts these effects and increases NAD+ levels. hSFRP2 mAb treatment further rescued the suppression of T-cell proliferation by tumor cells in a co-culture model. Finally, hSFRP2 mAb induced apoptosis in RF420 and RF577 OS cells but not in T-cells. Thus, hSFRP2 mAb therapy could potentially overcome PD-1 inhibitor resistance in metastatic osteosarcoma.
Collapse
Affiliation(s)
- Patrick Nasarre
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (P.N.); (D.I.G.); (J.B.S.); (I.V.B.); (R.M.); (E.H.); (P.C.); (M.L.); (A.A.J.); (S.M.)
| | - Denise I. Garcia
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (P.N.); (D.I.G.); (J.B.S.); (I.V.B.); (R.M.); (E.H.); (P.C.); (M.L.); (A.A.J.); (S.M.)
| | - Julie B. Siegel
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (P.N.); (D.I.G.); (J.B.S.); (I.V.B.); (R.M.); (E.H.); (P.C.); (M.L.); (A.A.J.); (S.M.)
| | - Ingrid V. Bonilla
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (P.N.); (D.I.G.); (J.B.S.); (I.V.B.); (R.M.); (E.H.); (P.C.); (M.L.); (A.A.J.); (S.M.)
| | - Rupak Mukherjee
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (P.N.); (D.I.G.); (J.B.S.); (I.V.B.); (R.M.); (E.H.); (P.C.); (M.L.); (A.A.J.); (S.M.)
| | - Eleanor Hilliard
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (P.N.); (D.I.G.); (J.B.S.); (I.V.B.); (R.M.); (E.H.); (P.C.); (M.L.); (A.A.J.); (S.M.)
| | - Paramita Chakraborty
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (P.N.); (D.I.G.); (J.B.S.); (I.V.B.); (R.M.); (E.H.); (P.C.); (M.L.); (A.A.J.); (S.M.)
| | - Cécile Nasarre
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Jason T. Yustein
- Department of Pediatrics, The Faris D. Virani Ewing Sarcoma Center at the Texas Children’s Cancer and Hematology Centers, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Margaret Lang
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (P.N.); (D.I.G.); (J.B.S.); (I.V.B.); (R.M.); (E.H.); (P.C.); (M.L.); (A.A.J.); (S.M.)
| | - Aneese A. Jaffa
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (P.N.); (D.I.G.); (J.B.S.); (I.V.B.); (R.M.); (E.H.); (P.C.); (M.L.); (A.A.J.); (S.M.)
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (P.N.); (D.I.G.); (J.B.S.); (I.V.B.); (R.M.); (E.H.); (P.C.); (M.L.); (A.A.J.); (S.M.)
| | - Nancy Klauber-DeMore
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (P.N.); (D.I.G.); (J.B.S.); (I.V.B.); (R.M.); (E.H.); (P.C.); (M.L.); (A.A.J.); (S.M.)
- Correspondence:
| |
Collapse
|
11
|
Syndecan-1 (CD138), Carcinomas and EMT. Int J Mol Sci 2021; 22:ijms22084227. [PMID: 33921767 PMCID: PMC8072910 DOI: 10.3390/ijms22084227] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Cell surface proteoglycans are known to be important regulators of many aspects of cell behavior. The principal family of transmembrane proteoglycans is the syndecans, of which there are four in mammals. Syndecan-1 is mostly restricted to epithelia, and bears heparan sulfate chains that are capable of interacting with a large array of polypeptides, including extracellular matrix components and potent mediators of proliferation, adhesion and migration. For this reason, it has been studied extensively with respect to carcinomas and tumor progression. Frequently, but not always, syndecan-1 levels decrease as tumor grade, stage and invasiveness and dedifferentiation increase. This parallels experiments that show depletion of syndecan-1 can be accompanied by loss of cadherin-mediated adhesion. However, in some tumors, levels of syndecan-1 increase, but the characterization of its distribution is relevant. There can be loss of membrane staining, but acquisition of cytoplasmic and/or nuclear staining that is abnormal. Moreover, the appearance of syndecan-1 in the tumor stroma, either associated with its cellular component or the collagenous matrix, is nearly always a sign of poor prognosis. Given its relevance to myeloma progression, syndecan-1-directed antibody—toxin conjugates are being tested in clinical and preclinical trials, and may have future relevance to some carcinomas.
Collapse
|
12
|
Belantamab Mafodotin to Treat Multiple Myeloma: A Comprehensive Review of Disease, Drug Efficacy and Side Effects. ACTA ACUST UNITED AC 2021; 28:640-660. [PMID: 33494319 PMCID: PMC7924384 DOI: 10.3390/curroncol28010063] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/09/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023]
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by excessive clonal proliferation of plasma cells. The treatment of multiple myeloma presents a variety of unique challenges due to the complex molecular pathophysiology and incurable status of the disease at this time. Given that MM is the second most common blood cancer with a characteristic and unavoidable relapse/refractory state during the course of the disease, the development of new therapeutic modalities is crucial. Belantamab mafodotin (belamaf, GSK2857916) is a first-in-class therapeutic, indicated for patients who have previously attempted four other treatments, including an anti-CD38 monoclonal antibody, a proteosome inhibitor, and an immunomodulatory agent. In November 2017, the FDA designated belamaf as a breakthrough therapy for heavily pretreated patients with relapsed/refractory multiple myeloma. In August 2020, the FDA granted accelerated approval as a monotherapy for relapsed or treatment-refractory multiple myeloma. The drug was also approved in the EU for this indication in late August 2020. Of note, belamaf is associated with the following adverse events: decreased platelets, corneal disease, decreased or blurred vision, anemia, infusion-related reactions, pyrexia, and fetal risk, among others. Further studies are necessary to evaluate efficacy in comparison to other standard treatment modalities and as future drugs in this class are developed.
Collapse
|
13
|
Richter J, Thibaud S. Anti-body building: The exercise of advancing immune based myeloma therapies. Blood Rev 2020; 48:100789. [PMID: 33384171 DOI: 10.1016/j.blre.2020.100789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/27/2020] [Accepted: 12/16/2020] [Indexed: 12/26/2022]
Abstract
The last decade has seen a marked improvement in the outcomes of patients with multiple myeloma. Much of this has been due to not only the advent of new therapies, but their inherent ability to be combined into 3 and 4 drug regimens without resulting in unacceptable toxicity. The general gestalt has been to combine agents of varied mechanisms of action. With the primary classes of agents such as proteasome inhibitors and immunomodulatory drugs as bases, the advent of antibody-based therapy in myeloma has allowed us to easily augment these therapies; much in the same way rituximab impacted the lymphoma world. With the approvals of daratumumab, elotuzumab and isatuximab; the myeloma world was ushered into the next wave of targeted agents. Here, we take a look at the current landscape of "off-the-shelf" antibody-based therapies in myeloma and peer into the next wave of multi-functional targeted agents.
Collapse
Affiliation(s)
- Joshua Richter
- Tisch Cancer Institute: Icahn School of Medicine at Mount Sinai, USA.
| | - Santiago Thibaud
- Tisch Cancer Institute: Icahn School of Medicine at Mount Sinai, USA.
| |
Collapse
|
14
|
Ghous G, Shoukat HMH, Tarar ZI, Zafar MU. A Review of Interstitial Pneumonitis Caused by Elotuzumab Through Case Discussions and Academic Writings. Cureus 2020; 12:e11162. [PMID: 33251069 PMCID: PMC7686952 DOI: 10.7759/cureus.11162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Patients with relapsed or refractory multiple myeloma have undergone two or three previous therapies are now being treated with a humanized IgG1 monoclonal antibody elotuzumab (HuLuc63) that targets F7 signalling lymphocytic activation molecule F7 (SLAMF7)- a signalling lymphocytic activation molecule. It is combined with dexamethasone and lenalidomide/pomalidomide for therapy. Adverse effects associated with elotuzumab consists of peripheral neuropathy, fever, constitutional symptoms (fatigue, headache, decreased appetite), and infections. A rare side effect of interstitial lung disease has only been observed in a single case. There are two case studies presented below of hypoxic respiratory failure upon this monoclonal antibody treatment both were successfully treated with steroid therapy. This article brings forth the hypothesis that elotuzumab can cause pneumonitis, and discontinuation of elotuzumab along with high-dose corticosteroids helps reverse the pneumonitis.
Collapse
|
15
|
Novel Approaches to Improve Myeloma Cell Killing by Monoclonal Antibodies. J Clin Med 2020; 9:jcm9092864. [PMID: 32899714 PMCID: PMC7564331 DOI: 10.3390/jcm9092864] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022] Open
Abstract
The monoclonal antibodies (mAbs) have significantly changed the treatment of multiple myeloma (MM) patients. However, despite their introduction, MM remains an incurable disease. The mAbs currently used for MM treatment were developed with different mechanisms of action able to target antigens, such as cluster of differentiation 38 (CD38) and SLAM family member 7 (SLAMF7) expressed by both, MM cells and the immune microenvironment cells. In this review, we focused on the mechanisms of action of the main mAbs approved for the therapy of MM, and on the possible novel approaches to improve MM cell killing by mAbs. Actually, the combination of anti-CD38 or anti-SLAMF7 mAbs with the immunomodulatory drugs significantly improved the clinical effect in MM patients. On the other hand, pre-clinical evidence indicates that different approaches may increase the efficacy of mAbs. The use of trans-retinoic acid, the cyclophosphamide or the combination of anti-CD47 and anti-CD137 mAbs have given the rationale to design these types of combinations therapies in MM patients in the future. In conclusion, a better understanding of the mechanism of action of the mAbs will allow us to develop novel therapeutic approaches to improve their response rate and to overcome their resistance in MM patients.
Collapse
|
16
|
Musto P, La Rocca F. Monoclonal antibodies in newly diagnosed and smoldering multiple myeloma: an updated review of current clinical evidence. Expert Rev Hematol 2020; 13:501-517. [PMID: 32290723 DOI: 10.1080/17474086.2020.1753502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Monoclonal antibodies (MoAbs) are rapidly changing the therapeutic scenario of multiple myeloma. Most of the available data, however, come from studies performed in patients with relapsed or refractory disease.Area covered: Here, the most recent results from clinical trials that have investigated (or are investigating) efficacy and safety of MoAbs as front-line treatments in both transplant-eligible and not-eligible patients with newly diagnosed multiple myeloma, as well as in smoldering myeloma, are reviewed. PubMed reported articles before 28 March 2020, and abstracts presented at the last ASCO, ASH, EHA, and IMW meetings were considered. Among others, pertinent data regarding daratumumab, isatuximab, elotuzumab, and pembrolizumab will be analyzed.Expert opinion: Introduction of MoAbs as first-line therapy will likely provide a significant improvement in the clinical outcome of patients with multiple myeloma. This will also require an appropriate re-positioning of salvage therapies. The role of MoAbs in smoldering myeloma appears to be promising, but adequate follow-up is needed.
Collapse
Affiliation(s)
- Pellegrino Musto
- Chair of Hematology and Unit of Hematology and Stem Cell Transplantation, "Aldo Moro" University School of Medicine, AOU Consorziale Policlinico, Bari, Italy
| | - Francesco La Rocca
- Laboratory of Advanced Diagnostics and Clinical Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (Pz), Italy
| |
Collapse
|