1
|
Mekala JR, Nalluri HP, Reddy PN, S B S, N S SK, G V S D SK, Dhiman R, Chamarthy S, Komaragiri RR, Manyam RR, Dirisala VR. Emerging trends and therapeutic applications of monoclonal antibodies. Gene 2024; 925:148607. [PMID: 38797505 DOI: 10.1016/j.gene.2024.148607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Monoclonal antibodies (mAbs) are being used to prevent, detect, and treat a broad spectrum of malignancies and infectious and autoimmune diseases. Over the past few years, the market for mAbs has grown exponentially. They have become a significant part of many pharmaceutical product lines, and more than 250 therapeutic mAbs are undergoing clinical trials. Ever since the advent of hybridoma technology, antibody-based therapeutics were realized using murine antibodies which further progressed into humanized and fully human antibodies, reducing the risk of immunogenicity. Some of the benefits of using mAbs over conventional drugs include a drastic reduction in the chances of adverse reactions, interactions between drugs, and targeting specific proteins. While antibodies are very efficient, their higher production costs impede the process of commercialization. However, their cost factor has been improved by developing biosimilar antibodies, which are affordable versions of therapeutic antibodies. Along with biosimilars, innovations in antibody engineering have helped to design bio-better antibodies with improved efficacy than the conventional ones. These novel mAb-based therapeutics are set to revolutionize existing drug therapies targeting a wide spectrum of diseases, thereby meeting several unmet medical needs. In the future, mAbs generated by applying next-generation sequencing (NGS) are expected to become a powerful tool in clinical therapeutics. This article describes the methods of mAb production, pre-clinical and clinical development of mAbs, approved indications targeted by mAbs, and novel developments in the field of mAb research.
Collapse
Affiliation(s)
- Janaki Ramaiah Mekala
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA.
| | - Hari P Nalluri
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Prakash Narayana Reddy
- Department of Microbiology, Dr. V.S. Krishna Government College, Visakhapatnam 530013, India
| | - Sainath S B
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524320, AP, India
| | - Sampath Kumar N S
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Sai Kiran G V S D
- Santhiram Medical College and General Hospital, Nandyal, Kurnool 518501, AP, India
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Sciences, National Institute of Technology Rourkela-769008, India
| | - Sahiti Chamarthy
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA
| | - Raghava Rao Komaragiri
- Department of CSE, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522302, Andhra Pradesh, INDIA
| | - Rajasekhar Reddy Manyam
- Amrita School of Computing, Amrita Vishwa Vidyapeetham, Amaravati Campus, Amaravati, Andhra Pradesh, India
| | - Vijaya R Dirisala
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India.
| |
Collapse
|
2
|
Paul S, Konig MF, Pardoll DM, Bettegowda C, Papadopoulos N, Wright KM, Gabelli SB, Ho M, van Elsas A, Zhou S. Cancer therapy with antibodies. Nat Rev Cancer 2024; 24:399-426. [PMID: 38740967 PMCID: PMC11180426 DOI: 10.1038/s41568-024-00690-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/29/2024] [Indexed: 05/16/2024]
Abstract
The greatest challenge in cancer therapy is to eradicate cancer cells with minimal damage to normal cells. Targeted therapy has been developed to meet that challenge, showing a substantially increased therapeutic index compared with conventional cancer therapies. Antibodies are important members of the family of targeted therapeutic agents because of their extraordinarily high specificity to the target antigens. Therapeutic antibodies use a range of mechanisms that directly or indirectly kill the cancer cells. Early antibodies were developed to directly antagonize targets on cancer cells. This was followed by advancements in linker technologies that allowed the production of antibody-drug conjugates (ADCs) that guide cytotoxic payloads to the cancer cells. Improvement in our understanding of the biology of T cells led to the production of immune checkpoint-inhibiting antibodies that indirectly kill the cancer cells through activation of the T cells. Even more recently, bispecific antibodies were synthetically designed to redirect the T cells of a patient to kill the cancer cells. In this Review, we summarize the different approaches used by therapeutic antibodies to target cancer cells. We discuss their mechanisms of action, the structural basis for target specificity, clinical applications and the ongoing research to improve efficacy and reduce toxicity.
Collapse
Affiliation(s)
- Suman Paul
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Maximilian F Konig
- Division of Rheumatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Katharine M Wright
- Discovery Chemistry, Merck Research Laboratory, Merck and Co, West Point, PA, USA
| | - Sandra B Gabelli
- Discovery Chemistry, Merck Research Laboratory, Merck and Co, West Point, PA, USA.
| | - Mitchell Ho
- Antibody Engineering Program, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | | | - Shibin Zhou
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Olivero G, Roggeri A, Pittaluga A. Anti-NMDA and Anti-AMPA Receptor Antibodies in Central Disorders: Preclinical Approaches to Assess Their Pathological Role and Translatability to Clinic. Int J Mol Sci 2023; 24:14905. [PMID: 37834353 PMCID: PMC10573896 DOI: 10.3390/ijms241914905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/30/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Autoantibodies against NMDA and AMPA receptors have been identified in the central nervous system of patients suffering from brain disorders characterized by neurological and psychiatric symptoms. It has been demonstrated that these autoantibodies can affect the functions and/or the expression of the targeted receptors, altering synaptic communication. The importance to clarify, in preclinical models, the molecular mechanisms involved in the autoantibody-mediated effects has emerged in order to understand their pathogenic role in central disorders, but also to propose new therapeutic approaches for preventing the deleterious central consequences. In this review, we describe some of the available preclinical literature concerning the impact of antibodies recognizing NMDA and AMPA receptors in neurons. This review discusses the cellular events that would support the detrimental roles of the autoantibodies, also illustrating some contrasting findings that in our opinion deserve attention and further investigations before translating the preclinical observations to clinic.
Collapse
Affiliation(s)
- Guendalina Olivero
- Department of Pharmacy (DiFar), University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (G.O.); (A.R.)
| | - Alessandra Roggeri
- Department of Pharmacy (DiFar), University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (G.O.); (A.R.)
| | - Anna Pittaluga
- Center of Excellence for Biomedical Research, 3Rs Center, Department of Pharmacy (DiFar), University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16145 Genoa, Italy
| |
Collapse
|
4
|
Advances in antibody-based therapy in oncology. NATURE CANCER 2023; 4:165-180. [PMID: 36806801 DOI: 10.1038/s43018-023-00516-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 01/10/2023] [Indexed: 02/22/2023]
Abstract
Monoclonal antibodies are a growing class of targeted cancer therapeutics, characterized by exquisite specificity, long serum half-life, high affinity and immune effector functions. In this review, we outline key advances in the field with a particular focus on recent and emerging classes of engineered antibody therapeutic candidates, discuss molecular structure and mechanisms of action and provide updates on clinical development and practice.
Collapse
|
5
|
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
|
6
|
Iżykowska K, Rassek K, Korsak D, Przybylski GK. Novel targeted therapies of T cell lymphomas. J Hematol Oncol 2020; 13:176. [PMID: 33384022 PMCID: PMC7775630 DOI: 10.1186/s13045-020-01006-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023] Open
Abstract
T cell lymphomas (TCL) comprise a heterogeneous group of non-Hodgkin lymphomas (NHL) that often present at an advanced stage at the time of diagnosis and that most commonly have an aggressive clinical course. Treatment in the front-line setting is most often cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) or CHOP-like regimens, which are effective in B cell lymphomas, but in TCL are associated with a high failure rate and frequent relapses. Furthermore, in contrast to B cell NHL, in which substantial clinical progress has been made with the introduction of monoclonal antibodies, no comparable advances have been seen in TCL. To change this situation and improve the prognosis in TCL, new gene-targeted therapies must be developed. This is now possible due to enormous progress that has been made in the last years in the understanding of the biology and molecular pathogenesis of TCL, which enables the implementation of the research findings in clinical practice. In this review, we present new therapies and current clinical and preclinical trials on targeted treatments for TCL using histone deacetylase inhibitors (HDACi), antibodies, chimeric antigen receptor T cells (CARTs), phosphatidylinositol 3-kinase inhibitors (PI3Ki), anaplastic lymphoma kinase inhibitors (ALKi), and antibiotics, used alone or in combinations. The recent clinical success of ALKi and conjugated anti-CD30 antibody (brentuximab-vedotin) suggests that novel therapies for TCL can significantly improve outcomes when properly targeted.
Collapse
Affiliation(s)
- Katarzyna Iżykowska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479, Poznań, Poland
| | - Karolina Rassek
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479, Poznań, Poland
| | - Dorota Korsak
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479, Poznań, Poland
| | - Grzegorz K Przybylski
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479, Poznań, Poland.
| |
Collapse
|
7
|
Wang L, Qin W, Huo YJ, Li X, Shi Q, Rasko JEJ, Janin A, Zhao WL. Advances in targeted therapy for malignant lymphoma. Signal Transduct Target Ther 2020; 5:15. [PMID: 32296035 PMCID: PMC7058622 DOI: 10.1038/s41392-020-0113-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/10/2019] [Accepted: 12/17/2019] [Indexed: 12/24/2022] Open
Abstract
The incidence of lymphoma has gradually increased over previous decades, and it ranks among the ten most prevalent cancers worldwide. With the development of targeted therapeutic strategies, though a subset of lymphoma patients has become curable, the treatment of refractory and relapsed diseases remains challenging. Many efforts have been made to explore new targets and to develop corresponding therapies. In addition to novel antibodies targeting surface antigens and small molecular inhibitors targeting oncogenic signaling pathways and tumor suppressors, immune checkpoint inhibitors and chimeric antigen receptor T-cells have been rapidly developed to target the tumor microenvironment. Although these targeted agents have shown great success in treating lymphoma patients, adverse events should be noted. The selection of the most suitable candidates, optimal dosage, and effective combinations warrant further investigation. In this review, we systematically outlined the advances in targeted therapy for malignant lymphoma, providing a clinical rationale for mechanism-based lymphoma treatment in the era of precision medicine.
Collapse
Affiliation(s)
- Li Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
| | - Wei Qin
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China
| | - Yu-Jia Huo
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China
| | - Xiao Li
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China
| | - Qing Shi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China
| | - John E J Rasko
- Gene & Stem Cell Therapy Program Centenary Institute, Sydney Medical School, University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Anne Janin
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
- U1165 Inserm/Université Paris 7, Hôpital Saint Louis, Paris, France
| | - Wei-Li Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China.
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.
| |
Collapse
|
8
|
Evan JR, Bozkurt SB, Thomas NC, Bagnato F. Alemtuzumab for the treatment of multiple sclerosis. Expert Opin Biol Ther 2018; 18:323-334. [PMID: 29309202 DOI: 10.1080/14712598.2018.1425388] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Alemtuzumab is a monoclonal antibody that targets for the destruction CD52+ cells, particularly B and T cells. Alemtuzumab is approved in more than 50 countries around the world for the treatment of adult patients with relapsing remitting multiple sclerosis (MS). Areas covered: In this review, the authors summarize biological, clinical and safety data related to the use of alemtuzumab in patients with MS. The authors then provide their expert opinion on alemtuzumab and the field as of whole before providing their perspectives for the future. Expert opinion: Alemtuzumab is highly efficacious; more so than first line treatments but comparable to natalizumab. Treatment schedule makes alemtuzumab administration easy and attractive to patients. However, its safety profile makes it a choice for a very limited number of patients, in a specific disease window. As of now, a cure for MS remains elusive and there is an unmet need for a safe and highly potent agent at the level of and beyond the blood brain barrier.
Collapse
Affiliation(s)
- Jennifer R Evan
- a Department of Neurology, Neuroimmunology Division/Neuroimaging Unit , Vanderbilt University Medical Center , Nashville , TN , USA.,b MedStar Health Union Memorial Hospital , Baltimore , MD , USA
| | - Subutay B Bozkurt
- a Department of Neurology, Neuroimmunology Division/Neuroimaging Unit , Vanderbilt University Medical Center , Nashville , TN , USA.,c University of Szeged Medical School , Szeged , Hungary
| | - Nikita C Thomas
- a Department of Neurology, Neuroimmunology Division/Neuroimaging Unit , Vanderbilt University Medical Center , Nashville , TN , USA
| | - Francesca Bagnato
- a Department of Neurology, Neuroimmunology Division/Neuroimaging Unit , Vanderbilt University Medical Center , Nashville , TN , USA
| |
Collapse
|
9
|
Villela-Ma LM, Velez-Ayal AK, Lopez-Sanc RDC, Martinez-C JA, Hernandez- JA. Advantages of Drug Selective Distribution in Cancer Treatment: Brentuximab Vedotin. INT J PHARMACOL 2017. [DOI: 10.3923/ijp.2017.785.807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
10
|
Poropatich K, Fontanarosa J, Samant S, Sosman JA, Zhang B. Cancer Immunotherapies: Are They as Effective in the Elderly? Drugs Aging 2017; 34:567-581. [DOI: 10.1007/s40266-017-0479-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
11
|
Sudulagunta SR, Kumbhat M, Sodalagunta MB, Settikere Nataraju A, Bangalore Raja SK, Thejaswi KC, Deepak R, Mohammed AH, Sunny SP, Visweswar A, Suvarna M, Nanjappa R. Warm Autoimmune Hemolytic Anemia: Clinical Profile and Management. J Hematol 2017; 6:12-20. [PMID: 32300386 PMCID: PMC7155818 DOI: 10.14740/jh303w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2017] [Indexed: 12/20/2022] Open
Abstract
Background Autoimmune hemolytic anemia (AIHA) is a rare autoimmune disease in which autoantibodies target red blood cells leading to marked decrease in their lifespan. The classification of AIHA is based on the immunochemical properties of the RBC autoantibody. Warm antibody AIHA (wAIHA) accounts for 75-80% of all adult AIHA cases. The treatment of wAIHA is mainly corticosteroids. Our retrospective study aimed to study the clinical profile and management of wAIHA. Methods Data of 75 patients admitted with wAIHA or presented to outpatient department (previous medical records) with wAIHA between January 2003 and January 2016 were analyzed. Results In our study, females constituted 12 and 26 patients of primary and secondary wAIHA, while males constituted 17 and 20 patients of primary and secondary wAIHA, respectively. Mean hemoglobin level at AIHA onset was found to be 7.1 ± 1.7 g/dL in primary wAIHA group and 6.3 ± 1.2 g/dL in secondary wAIHA group, which is statistically significant. Splenectomy was used as mode of treatment in one (3.4%) patient of primary wAIHA group and 15 (32.60%) patients of secondary wAIHA group, which is statistically significant. Mean age of wAIHA onset was 69.7 ± 21.5 years in wAIHA group secondary to lymphoma and 54.3 ± 25.7 years in other wAIHA group, which is statistically significant. Conclusion The most common causes of secondary wAIHA are B-cell lymphoma, systemic lupus erythematosus, rheumatoid arthritis, chronic lymphocytic leukemia (CLL), common variable immune deficiency, renal cell carcinoma and secondary to drug usage (alpha methyldopa and carbamazepine), respectively. Reducing the cumulative dose of corticosteroids with second line treatment whenever possible and therefore reducing the risk of sepsis, specifically in older patients with comorbidities will reduce morbidity and mortality.
Collapse
Affiliation(s)
| | - Monica Kumbhat
- Department of Pathology, Sri Ramachandra Medical College, Chennai, India
| | | | | | | | | | - Raj Deepak
- Department of General Medicine, Dr.B.R.Ambedkar Medical College, Bangalore, India
| | | | - Sony P Sunny
- Department of General Medicine, Dr.B.R.Ambedkar Medical College, Bangalore, India
| | - Amulya Visweswar
- Department of General Medicine, Dr.B.R.Ambedkar Medical College, Bangalore, India
| | - Mikita Suvarna
- Department of General Medicine, Dr.B.R.Ambedkar Medical College, Bangalore, India
| | - Rashmi Nanjappa
- Department of General Medicine, Dr.B.R.Ambedkar Medical College, Bangalore, India
| |
Collapse
|
12
|
Zhao Y, Su H, Shen X, Du J, Zhang X, Zhao Y. The immunological function of CD52 and its targeting in organ transplantation. Inflamm Res 2017; 66:571-578. [PMID: 28283679 DOI: 10.1007/s00011-017-1032-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 02/18/2017] [Accepted: 02/22/2017] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION CD52 (Campath-1 antigen), a glycoprotein of 12 amino acids anchored to glycosylphosphatidylinositol, is widely expressed on the cell surface of immune cells, such as mature lymphocytes, natural killer cells (NK), eosinophils, neutrophils, monocytes/macrophages, and dendritic cells (DCs). The anti-CD52 mAb, alemtuzumab, was used widely in clinics for the treatment of patients such as organ transplantation. In the present manuscript, we will briefly summarize the immunological function of CD52 and discuss the application of anti-CD52 mAb in transplantation settings. FINDINGS We reviewed studies published until July 2016 to explore the role of CD52 in immune cell function and its implication in organ transplantation. We showed that ligation of cell surface CD52 molecules may offer costimulatory signals for T-cell activation and proliferation. However, soluble CD52 molecules will interact with the inhibitory sialic acid-binding immunoglobulin-like lectin 10 (Siglec10) to significantly inhibit T cell proliferation and activation. Although the physiological and pathological significances of CD52 molecules are still poorly understood, the anti-CD52 mAb, alemtuzumab, was used widely for the treatment of patients with chronic lymphocytic leukemia, autoimmune diseases as well as cell and organ transplantation in clinics. CONCLUSION Studies clearly showed that CD52 can modulate T-cell activation either by its intracellular signal pathways or by the interaction of soluble CD52 and Siglec-10 expressing on T cells. However, the regulatory functions of CD52 on other immune cell subpopulations in organ transplantation require to be studied in the near future.
Collapse
Affiliation(s)
- Yang Zhao
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Huiting Su
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaofei Shen
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
- Department of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Junfeng Du
- Department of General Surgery, PLA Army General Hospital, Dongsishitiao Namencang 5, Dongcheng District, Beijing, 100007, China.
| | - Xiaodong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gong Ti Nan Road, Chaoyang District, Beijing, 100020, China.
| | - Yong Zhao
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
13
|
Thurgood LA, Chataway TK, Lower KM, Kuss BJ. From genome to proteome: Looking beyond DNA and RNA in chronic lymphocytic leukemia. J Proteomics 2017; 155:73-84. [PMID: 28069558 DOI: 10.1016/j.jprot.2017.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/11/2016] [Accepted: 01/03/2017] [Indexed: 02/07/2023]
Abstract
Chronic lymphocytic leukemia (CLL) remains the most common leukemia in the Western world. Whilst its disease course is extremely heterogeneous (ranging from indolent to aggressive), current methods are unable to accurately predict the clinical journey of each patient. There is clearly a pressing need for both improved prognostication and treatment options for patients with this disease. Whilst molecular studies have analyzed both genetic mutations and gene expression profiles of these malignant B-cells, and as a result have shed light on the pathogenesis of CLL, proteomic studies have been largely overlooked to date. This review summarizes our current knowledge of the proteomics of CLL, and discusses some of the issues in CLL proteomic research, such as reproducibility and data interpretation. In addition, we look ahead to how proteomics may significantly help in the development of a successful treatment for this currently incurable disease.
Collapse
Affiliation(s)
- Lauren A Thurgood
- Department of Haematology and Genetic Pathology, Flinders University, Adelaide, South Australia, Australia.
| | - Tim K Chataway
- Department of Physiology, Flinders University, Adelaide, South Australia, Australia
| | - Karen M Lower
- Department of Haematology and Genetic Pathology, Flinders University, Adelaide, South Australia, Australia
| | - Bryone J Kuss
- Department of Haematology and Genetic Pathology, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
14
|
Abstract
Immunosuppression strategies that selectively inhibit effector T cells while preserving and even enhancing CD4FOXP3 regulatory T cells (Treg) permit immune self-regulation and may allow minimization of immunosuppression and associated toxicities. Many immunosuppressive drugs were developed before the identity and function of Treg were appreciated. A good understanding of the interactions between Treg and immunosuppressive agents will be valuable to the effective design of more tolerable immunosuppression regimens. This review will discuss preclinical and clinical evidence regarding the influence of current and emerging immunosuppressive drugs on Treg homeostasis, stability, and function as a guideline for the selection and development of Treg-friendly immunosuppressive regimens.
Collapse
Affiliation(s)
- Akiko Furukawa
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Steven A Wisel
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
15
|
Gross CC, Ahmetspahic D, Ruck T, Schulte-Mecklenbeck A, Schwarte K, Jörgens S, Scheu S, Windhagen S, Graefe B, Melzer N, Klotz L, Arolt V, Wiendl H, Meuth SG, Alferink J. Alemtuzumab treatment alters circulating innate immune cells in multiple sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2016; 3:e289. [PMID: 27766281 PMCID: PMC5063395 DOI: 10.1212/nxi.0000000000000289] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/12/2016] [Indexed: 11/15/2022]
Abstract
Objective: To characterize changes in myeloid and lymphoid innate immune cells in patients with relapsing-remitting multiple sclerosis (MS) during a 6-month follow-up after alemtuzumab treatment. Methods: Circulating innate immune cells including myeloid cells and innate lymphoid cells (ILCs) were analyzed before and 6 and 12 months after onset of alemtuzumab treatment. Furthermore, a potential effect on granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)–23 production by myeloid cells and natural killer (NK) cell cytolytic activity was determined. Results: In comparison to CD4+ T lymphocytes, myeloid and lymphoid innate cell subsets of patients with MS expressed significantly lower amounts of CD52 on their cell surface. Six months after CD52 depletion, numbers of circulating plasmacytoid dendritic cells (DCs) and conventional DCs were reduced compared to baseline. GM-CSF and IL-23 production in DCs remained unchanged. Within the ILC compartment, the subset of CD56bright NK cells specifically expanded under alemtuzumab treatment, but their cytolytic activity did not change. Conclusions: Our findings demonstrate that 6 months after alemtuzumab treatment, specific DC subsets are reduced, while CD56bright NK cells expanded in patients with MS. Thus, alemtuzumab specifically restricts the DC compartment and expands the CD56bright NK cell subset with potential immunoregulatory properties in MS. We suggest that remodeling of the innate immune compartment may promote long-term efficacy of alemtuzumab and preserve immunocompetence in patients with MS.
Collapse
Affiliation(s)
- Catharina C Gross
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Diana Ahmetspahic
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Tobias Ruck
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Andreas Schulte-Mecklenbeck
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Kathrin Schwarte
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Silke Jörgens
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Stefanie Scheu
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Susanne Windhagen
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Bettina Graefe
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Nico Melzer
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Luisa Klotz
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Volker Arolt
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Heinz Wiendl
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Sven G Meuth
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Judith Alferink
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| |
Collapse
|
16
|
Abstract
Chronic lymphocytic leukaemia (CLL) is well known to generate impaired immune responses in the host, with the malignant clone residing in well-vascularized tissues and circulating in peripheral blood but also in close proximity to effector cells that are capable, if activated appropriately, of eliciting a cytotoxic response. These, combined with the fact that this is frequently a condition affecting older patients with co-morbidities often unfit for many "traditional" cytotoxic agents with their significant associated toxicities, make CLL an ideal candidate for the development of immunotherapy. The impressive results seen with the addition of a monoclonal antibody, rituximab, to a chemotherapy backbone, for example, is testament to how effective harnessing an immune-mediated response in CLL can be. This review serves to outline the available arsenal of immunotherapies-past and present-demonstrated to have potential in CLL with some perspectives on how the landscape in this disease may evolve in the future.
Collapse
Affiliation(s)
- Ciara L. Freeman
- John Vane Cancer Centre, Charterhouse Square, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - John G. Gribben
- John Vane Cancer Centre, Charterhouse Square, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
17
|
Alemtuzumab and CHOP Chemotherapy for the Treatment of Aggressive Histology Peripheral T Cell Lymphomas: A Multi-Center Phase I Study. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2016; 16:18-28.e4. [DOI: 10.1016/j.clml.2015.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 10/01/2015] [Accepted: 11/12/2015] [Indexed: 02/02/2023]
|
18
|
Holgate RGE, Weldon R, Jones TD, Baker MP. Characterisation of a Novel Anti-CD52 Antibody with Improved Efficacy and Reduced Immunogenicity. PLoS One 2015; 10:e0138123. [PMID: 26372145 PMCID: PMC4570798 DOI: 10.1371/journal.pone.0138123] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/25/2015] [Indexed: 02/02/2023] Open
Abstract
Anti-CD52 therapy has been shown to be effective in the treatment of a number of B cell malignancies, hematopoietic disorders and autoimmune diseases (including rheumatoid arthritis and multiple sclerosis); however the current standard of treatment, the humanized monoclonal antibody alemtuzumab, is associated with the development of anti-drug antibodies in a high proportion of patients. In order to address this problem, we have identified a novel murine anti-CD52 antibody which has been humanized using a process that avoids the inclusion within the variable domains of non-human germline MHC class II binding peptides and known CD4+ T cell epitopes, thus reducing its potential for immunogenicity in the clinic. The resultant humanized antibody, ANT1034, was shown to have superior binding to CD52 expressing cells than alemtuzumab and was more effective at directing both antibody dependent and complement dependent cell cytotoxicity. Furthermore, when in the presence of a cross-linking antibody, ANT1034 was more effective at directly inducing apoptosis than alemtuzumab. ANT1034 also showed superior activity in a SCID mouse/human CD52 tumour xenograft model where a single 1 mg/Kg dose of ANT1034 led to increased mouse survival compared to a 10 mg/Kg dose of alemtuzumab. Finally, ANT1034 was compared to alemtuzumab in in vitro T cell assays in order to evaluate its potential to stimulate proliferation of T cells in peripheral blood mononuclear cells derived from a panel of human donors: whereas alemtuzumab stimulated proliferation in a high proportion of the donor cohort, ANT1034 did not stimulate proliferation in any of the donors. Therefore we have developed a candidate therapeutic humanized antibody, ANT1034, that may have the potential to be more efficacious and less immunogenic than the current standard anti-CD52 therapy.
Collapse
Affiliation(s)
| | - Richard Weldon
- Antitope Limited, Babraham Research Campus, Cambridge, United Kingdom
| | - Timothy D. Jones
- Antitope Limited, Babraham Research Campus, Cambridge, United Kingdom
| | - Matthew P. Baker
- Antitope Limited, Babraham Research Campus, Cambridge, United Kingdom
| |
Collapse
|
19
|
A review of monoclonal antibody therapies in lymphoma. Crit Rev Oncol Hematol 2015; 97:72-84. [PMID: 26318093 DOI: 10.1016/j.critrevonc.2015.08.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 07/03/2015] [Accepted: 08/05/2015] [Indexed: 12/27/2022] Open
Abstract
Monoclonal antibodies (moAb) represent a novel way of delivering therapy through specific target antigens expressed on lymphoma cells and minimizes the collateral damage that is common with conventional chemotherapy. The paradigm of this approach is the targeting of CD20 by rituximab. Since its FDA approval in 1997, rituximab has become the standard of care in almost every line of therapy in most B-cell lymphomas. This review will briefly highlight some of the key rituximab trials while looking more closely at the evidence that is bringing other antibodies, including next generation anti-CD20 moAbs, and anti-CD30 moAbs, among others to the forefront of lymphoma therapy.
Collapse
|
20
|
Merli M, Ferrario A, Maffioli M, Arcaini L, Passamonti F. Investigational therapies targeting lymphocyte antigens for the treatment of non-Hodgkin's lymphoma. Expert Opin Investig Drugs 2015; 24:897-912. [PMID: 25900401 DOI: 10.1517/13543784.2015.1038342] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION The advent of the anti-CD20 mAb rituximab has opened a new era in the treatment of non-Hodgkin's lymphomas (NHL), markedly altering standard treatment strategies. Moreover, the proof-of-concept that targeting a specific lymphocyte surface antigen may induce a highly effective and safe targeted killing of malignant cells has opened the door to the development of a plethora of novel mAbs directed towards different B- and T-cell-specific antigens. AREAS COVERED This review discusses the recent available clinical data about new-generation anti-CD20 mAbs characterized by increased antibody- (obinutuzumab) or complement-dependent cyotoxicity (ofatumumab) as well as novel investigational agents targeting other lymphocyte antigens (e.g., CD19, CD22, CD30, CD40, CD52, CCR4), which are currently under investigation for B- and T-cell NHL treatment. In addition, antibody-drug conjugates (inotuzumab ozogamicin, brentuximab vedotin, polatuzumab vedotin), bispecific T-cell engagers (blinatumomab) and a new class of antibodies targeting cytotoxic T-lymphocyte-associated antigen 4, programmed death 1 or programmed death ligand 1 (immune checkpoint inhibitors) are specifically considered. EXPERT OPINION Among the novel mAbs challenging rituximab, obinutuzumab seems to be in the most advanced phase, with the results of randomized trials awaited shortly. Brentuximab vedotin is increasing its role in T-cell NHL. Furthermore, immune checkpoint inhibitors have the potential to acquire a great relevance in NHL therapy.
Collapse
Affiliation(s)
- Michele Merli
- University Hospital Ospedale di Circolo and Fondazione Macchi, Division of Hematology , Viale L. Borri 57, 21100 Varese , Italy +39 332 393 648 ;
| | | | | | | | | |
Collapse
|
21
|
Cannon AC, Loberiza FR. Review of Antibody-Based Immunotherapy in the Treatment of Non-Hodgkin Lymphoma and Patterns of Use. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2015; 15:129-38. [DOI: 10.1016/j.clml.2014.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/22/2014] [Accepted: 07/29/2014] [Indexed: 01/22/2023]
|
22
|
Dubey D, Cano CA, Stuve O. Intractable and highly active relapsing multiple sclerosis - role of alemtuzumab. Neuropsychiatr Dis Treat 2015; 11:2405-14. [PMID: 26425095 PMCID: PMC4581781 DOI: 10.2147/ndt.s90473] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Alemtuzumab is a humanized recombinant monoclonal antibody that was recently approved by the US Food and Drug Administration and the European Medicines Agency for the management of relapsing forms of multiple sclerosis (MS). It has been utilized for the management of chronic lymphocytic leukemia, bone marrow and renal transplantation, or graft versus host disease. Because of its immunomodulatory properties, it was brought into clinical development in MS. One Phase II (CAMMS223) and two Phase III clinical trials (CARE-MSI and -II) have evaluated the safety and efficacy of alemtuzumab in patients with relapsing-remitting MS. Even though its efficacy profile and long-lasting effect have attracted much interest among physicians and patients, it has significant potential adverse effects that may limit its use to patients with active disease. Here, we review the history of drug development of alemtuzumab. Furthermore, we outline the postulated mechanisms of action, clinical evidence, and safety of alemtuzumab for its use as a disease-modifying agent in active and highly active MS.
Collapse
Affiliation(s)
- Divyanshu Dubey
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher A Cano
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Olaf Stuve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA ; Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA ; Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
23
|
Caspase-8 polymorphisms result in reduced Alemtuzumab-induced T-cell apoptosis and worse survival after transplantation. Bone Marrow Transplant 2014; 50:237-43. [PMID: 25347010 DOI: 10.1038/bmt.2014.238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/11/2014] [Accepted: 09/13/2014] [Indexed: 11/08/2022]
Abstract
Allo-SCT using unrelated donors is a curative treatment for patients with hematological disorders. The best donor is one matched for 10/10 HLA alleles, however studies have shown an additional survival benefit when considering other genetic factors. It has been shown that a six-nucleotide insertion/deletion polymorphism in the CASP8 gene promoter results in reduced susceptibility of T lymphocytes to undergo apoptosis. In 186 SCT recipients, we found a significantly better OS in those who received a transplant from a WT/WT donor compared with donors with a deletion (3 years: 52 vs 34%; P=0.03; multivariate analysis; RR 0.61; 95% CI 0.38-0.98, P=0.04). This was more marked when both the patient and the donor had a deletion (3 years OS: 62% compared with 36%, P=0.01). As the majority of these patients received Alemtuzumab during conditioning, we went on to analyze the in vitro effect of the polymorphism on Alemtuzumab-induced apoptosis. We showed statistically significantly higher percentages of apoptotic naïve CD4 (P<0.0005) and CD8 (P<0.0005) T cells in WT/WT donors in comparison with donors with a deletion. These data imply an unrecognized role for the CASP8 promoter polymorphism on survival following unrelated SCT particularly in the context of T-cell depletion with Alemtuzumab.
Collapse
|
24
|
Suresh T, Lee LX, Joshi J, Barta SK. New antibody approaches to lymphoma therapy. J Hematol Oncol 2014; 7:58. [PMID: 25355407 PMCID: PMC4172963 DOI: 10.1186/s13045-014-0058-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/29/2014] [Indexed: 01/01/2023] Open
Abstract
The CD20-directed monoclonal antibody rituximab established a new era in lymphoma therapy. Since then other epitopes on the lymphoma surface have been identified as potential targets for monoclonal antibodies (mAb). While most mAbs eliminate lymphoma cells mainly by antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity or direct cell death, others counter mechanisms utilized by malignant cells to evade immune surveillance. Expression of PD-L1 on malignant or stromal cells in the tumor environment for example leads to T-cell anergy. Targeting either PD-1 or PD-L1 via mAbs can indirectly eliminate cancer cells by unblocking the host intrinsic immune response. Yet another mechanism of targeted therapy with mAbs are bi-specific T-cell engagers (BiTE) such as blinatumomab, which directly engages the host immune cells. These examples highlight the broad spectrum of available therapies targeting the lymphoma surface with mAbs utilizing both passive and active immune pathways. Many of these agents have already demonstrated significant activity in clinical trials. In this review we will focus on novel CD20-directed antibodies as well as mAbs directed against newer targets like CD19, CD22, CD40, CD52 and CCR4. In addition we will review mAbs unblocking immune checkpoints and the BiTE blinatumomab. Given the success of mAbs and the expansion in active and passive immunotherapies, these agents will play an increasing role in the treatment of lymphomas.
Collapse
|
25
|
Pevna M, Doubek M, Coupek P, Stehlikova O, Klabusay M. Residual cancer lymphocytes in patients with chronic lymphocytic leukemia after therapy show increased expression of surface antigen CD52 detected using quantitative fluorescence cytometry. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2014; 14:411-8. [PMID: 25066039 DOI: 10.1016/j.clml.2014.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 05/09/2014] [Accepted: 06/04/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Rituximab and alemtuzumab, mAbs used in recent years to treat CLL, are directed against antigens CD20 and CD52. CD20 is not highly expressed by CLL tumor cells, and rituximab does not have significant effectiveness in CLL unless combined with chemotherapy. Alemtuzumab targets CD52, which is much more highly expressed, and is currently the most effective agent used alone for CLL. Variability in expression of both antigens among these patients might be related to different individual therapeutic responses to mAb therapy. PATIENTS AND METHODS A total 95 patients diagnosed with CLL and/or SLL were divided into 4 groups: (1) untreated; (2) in complete or partial remission; (3) disease in progression; and (4) diagnosed with SLL. Flow cytometry of peripheral blood cells included gating of the CD5(+)CD19(+) tumor population, within which mean fluorescence intensity of fluorescein isothiocyanate (FITC) conjugated with anti-CD20 or anti-CD52 antibody was measured. The resulting expression of the 2 antigens was deduced from the calibration curve using Quantum FITC particles. RESULTS Expression of CD20 showed no significant differences among the 4 groups of patients. However, significantly greater expression of surface antigen CD52 was recorded in patient group 2 in complete or partial remission (P < .001). CONCLUSION The residual population of CLL cells after therapy is characterized by increased surface detection of CD52. Although the exact cause of this phenomenon is unknown, our results provide a basis to consider the potential for CLL consolidation therapy using alemtuzumab.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Alemtuzumab
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antigens, CD/blood
- Antigens, Neoplasm/blood
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- CD52 Antigen
- Calibration
- Female
- Flow Cytometry/methods
- Fluorescein-5-isothiocyanate/analysis
- Fluorescent Dyes/analysis
- Fluorometry/methods
- Glycoproteins/blood
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/blood
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Lymphocytes/chemistry
- Male
- Middle Aged
- Molecular Targeted Therapy
- Neoplasm, Residual
- Patient Selection
- Remission Induction
Collapse
Affiliation(s)
- Michaela Pevna
- International Clinical Research Center-Integrated Center of Cellular Therapy and Regenerative Medicine, St Anne's University Hospital Brno, Brno, Czech Republic
| | - Michael Doubek
- Department of Internal Medicine-Hematooncology, University Hospital Brno, Brno, Czech Republic
| | - Petr Coupek
- International Clinical Research Center-Integrated Center of Cellular Therapy and Regenerative Medicine, St Anne's University Hospital Brno, Brno, Czech Republic
| | - Olga Stehlikova
- Department of Internal Medicine-Hematooncology, University Hospital Brno, Brno, Czech Republic
| | - Martin Klabusay
- International Clinical Research Center-Integrated Center of Cellular Therapy and Regenerative Medicine, St Anne's University Hospital Brno, Brno, Czech Republic.
| |
Collapse
|
26
|
Lee F, Luevano M, Veys P, Yong K, Madrigal A, Shaw BE, Saudemont A. The effects of CAMPATH-1H on cell viability do not correlate to the CD52 density on the cell surface. PLoS One 2014; 9:e103254. [PMID: 25050704 PMCID: PMC4106894 DOI: 10.1371/journal.pone.0103254] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 06/30/2014] [Indexed: 11/23/2022] Open
Abstract
Graft versus host disease (GvHD) is one of the main complications after hematological stem cell transplantation (HSCT). CAMPATH-1H is used in the pre-transplant conditioning regimen to effectively reduce GvHD by targeting CD52 antigens on T cells resulting in their depletion. Information regarding CD52 expression and the effects of CAMPATH-1H on immune cells is scant and limited to peripheral blood (PB) T and B cells. To date, the effects of CAMPATH-1H on cord blood (CB) cells has not been studied. Here we aimed to analyze CD52 expression and the effects of CAMPATH-1H on fresh or frozen, resting or activated, PB mononuclear cells (PBMC) and CB mononuclear cells (CBMC). In resting state, CD52 expression was higher in CB than PB T cell subsets (653.66±26.68 vs 453.32±19.2) and B cells (622.2±20.65 vs 612.0±9.101) except for natural killer (NK) cells where CD52 levels were higher in PB (421.0±9.857) than CB (334.3±9.559). In contrast, CD52 levels were comparable across all cell types after activation. CAMPATH-1H depleted resting cells more effectively than activated cells with approximately 80–95% of apoptosis observed with low levels of necrosis. There was no direct correlation between cell surface CD52 density and depleting effects of CAMPATH-1H. In addition, no difference in cell viability was noted when different concentrations of CAMPATH-1H were used. CD52 was not expressed on HSC but began to be expressed as the cells differentiate, implying that CAMPATH-1H could potentially affect HSC differentiation and proliferation. Our study provides insightful information, which contributes to the better understanding in the use of CAMPATH-1H as part of the conditioning regime in HSCT.
Collapse
MESH Headings
- Alemtuzumab
- Antibodies, Monoclonal, Humanized/pharmacology
- Antigens, CD/analysis
- Antigens, CD/immunology
- Antigens, Neoplasm/analysis
- Antigens, Neoplasm/immunology
- Antineoplastic Agents/pharmacology
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- CD52 Antigen
- Cell Separation
- Cell Survival/drug effects
- Glycoproteins/analysis
- Glycoproteins/immunology
- Humans
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
Collapse
Affiliation(s)
- Fuiyee Lee
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
| | - Martha Luevano
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
| | - Paul Veys
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
- Great Ormond Street Hospital for Children NHS, London, United Kingdom
| | - Kwee Yong
- University College London, Cancer Institute, London, United Kingdom
| | - Alejandro Madrigal
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
| | - Bronwen E. Shaw
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
- Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Aurore Saudemont
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
- * E-mail:
| |
Collapse
|
27
|
D'Arena G, Laurenti L. Alemtuzumab and treatment of chronic lymphocytic leukemia and its immune-related disorders: one player on two tables. Acta Haematol 2014; 132:237-9. [PMID: 24853680 DOI: 10.1159/000359949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 01/23/2014] [Indexed: 11/19/2022]
|
28
|
Hoermann G, Blatt K, Greiner G, Putz EM, Berger A, Herrmann H, Cerny-Reiterer S, Gleixner KV, Walz C, Hoetzenecker K, Müllauer L, Reiter A, Sotlar K, Sexl V, Valent P, Mayerhofer M. CD52 is a molecular target in advanced systemic mastocytosis. FASEB J 2014; 28:3540-51. [PMID: 24760752 DOI: 10.1096/fj.14-250894] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Advanced systemic mastocytosis (SM) is an aggressive hematopoietic neoplasm with poor prognosis and short survival times. So far, no curative therapy is available for affected patients. We have identified the cell surface antigen CD52 (CAMPATH-1) as a molecular target expressed abundantly on the surface of primary neoplastic mast cells (MCs) in patients with advanced SM. In contrast, neoplastic MCs of patients with indolent SM and normal MCs expressed only low levels or did not express CD52. To study the mechanisms of CD52 expression and the value of this antigen as a potential therapeutic target, we generated a human MC cell line, designated MCPV-1, by lentiviral immortalization of cord blood-derived MC progenitor cells. Functional studies revealed that activated RAS profoundly promotes surface expression of CD52. The CD52-targeting antibody alemtuzumab induced cell death in CD52(+) primary neoplastic MCs obtained from patients with SM as well as in MCPV-1 cells. NSG mice xenotransplanted with MCPV-1 cells survived significantly longer after treatment with alemtuzumab (median survival: 31 d untreated vs. 46 d treated; P=0.0012). We conclude that CD52 is a novel marker and potential therapeutic target in neoplastic MCs in patients with advanced SM.
Collapse
Affiliation(s)
| | - Katharina Blatt
- Department of Internal Medicine I, Division of Hematology and Hemostaseology
| | | | - Eva Maria Putz
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Angelika Berger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Sabine Cerny-Reiterer
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Ludwig Boltzmann Cluster Oncology, Vienna, Austria
| | - Karoline V Gleixner
- Department of Internal Medicine I, Division of Hematology and Hemostaseology
| | - Christoph Walz
- Institute of Pathology, Ludwig Maximilians University Munich, Munich, Germany
| | | | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Andreas Reiter
- Medizinische Universitätsklinik, Universitätsmedizin Mannheim, Mannheim, Germany; and
| | - Karl Sotlar
- Institute of Pathology, Ludwig Maximilians University Munich, Munich, Germany
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Ludwig Boltzmann Cluster Oncology, Vienna, Austria
| | | |
Collapse
|
29
|
Ferrajoli A, Faderl S, Keating MJ. Monoclonal antibodies in chronic lymphocytic leukemia. Expert Rev Anticancer Ther 2014; 6:1231-8. [PMID: 17020457 DOI: 10.1586/14737140.6.9.1231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Multiple options are now available for the treatment of chronic lymphocytic leukemia. Over the last 10 years, monoclonal antibodies have become an integral part of the management of this disease. Alemtuzumab has received approval for use in patients with fludarabine-refractory chronic lymphocytic leukemia. Rituximab has been investigated extensively in chronic lymphocytic leukemia both as a single agent and in combination with chemotherapy and other monoclonal antibodies. Epratuzumab and lumiliximab are newer monoclonal antibodies in the early phase of clinical development. This article will review the monoclonal antibodies more commonly used to treat chronic lymphocytic leukemia, the results obtained with monoclonal antibodies as single agents and in combination with chemotherapy, and other biological agents and newer compounds undergoing clinical trials.
Collapse
MESH Headings
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
Collapse
Affiliation(s)
- Alessandra Ferrajoli
- University of Texas MD Anderson Cancer Center, Department of Leukemia, Unit 428, PO Box 301402, Houston, TX 77230-1402, USA.
| | | | | |
Collapse
|
30
|
|
31
|
Abstract
Immunoglobulins (Ig) or antibodies are heavy plasma proteins, with sugar chains added to amino-acid residues by N-linked glycosylation and occasionally by O-linked glycosylation. The versatility of antibodies is demonstrated by the various functions that they mediate such as neutralization, agglutination, fixation with activation of complement and activation of effector cells. Naturally occurring antibodies protect the organism against harmful pathogens, viruses and infections. In addition, almost any organic chemical induces antibody production of antibodies that would bind specifically to the chemical. These antibodies are often produced from multiple B cell clones and referred to as polyclonal antibodies. In recent years, scientists have exploited the highly evolved machinery of the immune system to produce structurally and functionally complex molecules such as antibodies from a single B clone, heralding the era of monoclonal antibodies. Most of the antibodies currently in the clinic, target components of the immune system, are not curative and seek to alleviate symptoms rather than cure disease. Our group used a novel strategy to identify reparative human monoclonal antibodies distinct from conventional antibodies. In this chapter, we discuss the therapeutic relevance of both polyclonal and monoclonal antibodies in clinic.
Collapse
Affiliation(s)
- Bharath Wootla
- Departments of Neurology and Immunology, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
32
|
Novitzky N, Davison G, Abdulla R, Mowla S. Definition of the Variables Affecting Efficacy of Immunodepletion Ex Vivo of Peripheral Blood Progenitor Cell Grafts by Alemtuzumab (Campath in the Bag). Biol Blood Marrow Transplant 2013; 19:1753-9. [DOI: 10.1016/j.bbmt.2013.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 10/01/2013] [Indexed: 11/25/2022]
|
33
|
Immunotherapy treatments of warm autoimmune hemolytic anemia. Clin Dev Immunol 2013; 2013:561852. [PMID: 24106518 PMCID: PMC3784078 DOI: 10.1155/2013/561852] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 07/31/2013] [Accepted: 08/13/2013] [Indexed: 12/31/2022]
Abstract
Warm autoimmune hemolytic anemia (WAIHA) is one of four clinical types of autoimmune hemolytic anemia (AIHA), with the characteristics of autoantibodies maximally active at body temperature. It produces a variable anemia—sometimes mild and sometimes severe. With respect to the absence or presence of an underlying condition, WAIHA is either idiopathic (primary) or secondary, which determines the treatment strategies in practice. Conventional treatments include immune suppression with corticosteroids and, in some cases, splenectomy. In recent years, the number of clinical studies with monoclonal antibodies and immunosuppressants in the treatment of WAIHA increased as the knowledge of autoimmunity mechanisms extended. This thread of developing new tools of treating WAIHA is well exemplified with the success in using anti-CD20 monoclonal antibody, Rituximab. Following this success, other treatment methods based on the immune mechanisms of WAIHA have emerged. We reviewed these newly developed immunotherapy treatments here in order to provide the clinicians with more options in selecting the best therapy for patients with WAIHA, hoping to stimulate researchers to find more novel immunotherapy strategies.
Collapse
|
34
|
Nastoupil LJ, Sinha R, Flowers CR. The role of chemotherapy in managing chronic lymphocytic leukemia: optimizing combinations with targeted therapy. Expert Rev Anticancer Ther 2013; 13:1089-108. [PMID: 23919536 DOI: 10.1586/14737140.2013.818294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
For many years, alkylating agents were the standard treatment for chronic lymphocytic leukemia (CLL). The advent of purine analogs improved response rates, but not overall survival, and although the monoclonal antibody rituximab is generally active against B-cell malignancies, it has demonstrated limited benefits as monotherapy for the treatment of CLL. However, specific combinations of chemotherapy, antibodies and targeted therapies have demonstrated additive or synergistic activity in CLL cells and deliver substantial clinical benefits. A greater understanding of the actions of chemotherapies and targeted agents on cellular pathways will advance the development of rationally designed combinations corresponding to individual patients' disease profiles.
Collapse
Affiliation(s)
- Loretta J Nastoupil
- Department of Hematology Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
35
|
Warner JL, Arnason JE. Alemtuzumab use in relapsed and refractory chronic lymphocytic leukemia: a history and discussion of future rational use. Ther Adv Hematol 2013; 3:375-89. [PMID: 23606939 DOI: 10.1177/2040620712458949] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In this review, we outline the clinical experience with single-agent alemtuzumab as a treatment for relapsed and refractory chronic lymphocytic leukemia (CLL) in both prospective and retrospective trials and describe the multiagent use of the drug with the goal of updating clinicians on recent developments and possible future rational combinations. Alemtuzumab, an antibody targeting the lymphocyte-specific surface marker CD52, is an approved agent for the treatment of CLL. Despite its demonstrated efficacy, likely secondary to concerns regarding infectious complications, it is most commonly used in the relapsed and refractory setting. Given alemtuzumab's unique mechanism of action it has been demonstrated to have activity in disease that is refractory to both alkylating agents and purine analogs. Furthermore, it has activity in TP53-mutated disease, which has the worst prognosis of any subset of CLL. Alemtuzumab has greater efficacy on circulating disease relative to nodal disease. Rational combinations are attempting to use these attributes to increase response rates in patients with relapsed and refractory disease.
Collapse
Affiliation(s)
- Jeremy L Warner
- Hematologic Malignancy and Bone Marrow Transplantation Program, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
36
|
Laurenti L, Vannata B, Innocenti I, Autore F, Santini F, Sica S, Efremov DG. The use of monoclonal antibodies in the treatment of autoimmune complications of chronic lymphocytic leukemia. Mediterr J Hematol Infect Dis 2013; 5:e2013027. [PMID: 23667725 PMCID: PMC3647707 DOI: 10.4084/mjhid.2013.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 04/04/2013] [Indexed: 12/27/2022] Open
Abstract
Autoimmune cytopenias are a frequent complication in CLL, occurring in approximately 5-10% of the patients. The most common manifestation is autoimmune haemolytic anaemia, followed by immune thrombocytopenia and only rarely pure red blood cell aplasia or autoimmune granulocytopenia. Initial treatment is as for the idiopathic autoimmune cytopenias, with most patients responding to conventional corticosteroid therapy. Patients, who do not respond to conventional therapy after 4-6 weeks, should be considered for alternative immunosuppression, monoclonal antibody therapy or splenectomy. While randomized trials demonstrating the benefit of rituximab in CLL-related autoimmune diseases are still lacking, there are considerable data in the literature that provide evidence for its effectiveness. The monoclonal antibody alemtuzumab also displays considerable activity against both the malignant disease and the autoimmune complication in patients with CLL, although at the expense of greater toxicity. A number of new monoclonal antibodies, such as ofatumumab, GA-101, lumiliximab, TRU-016, epratuzumab, and galiximab, are currently investigated in CLL and their activity in CLL-related autoimmune cytopenias should be evaluated in future studies.
Collapse
Affiliation(s)
- Luca Laurenti
- Department of Hematology, Catholic University of Rome, “A. Gemelli” Hospital, Largo A. Gemelli 8, Rome, Italy
| | - Barbara Vannata
- Department of Hematology, Catholic University of Rome, “A. Gemelli” Hospital, Largo A. Gemelli 8, Rome, Italy
| | - Idanna Innocenti
- Department of Hematology, Catholic University of Rome, “A. Gemelli” Hospital, Largo A. Gemelli 8, Rome, Italy
| | - Francesco Autore
- Department of Hematology, Catholic University of Rome, “A. Gemelli” Hospital, Largo A. Gemelli 8, Rome, Italy
| | - Francesco Santini
- Department of Hematology, Catholic University of Rome, “A. Gemelli” Hospital, Largo A. Gemelli 8, Rome, Italy
| | - Simona Sica
- Department of Hematology, Catholic University of Rome, “A. Gemelli” Hospital, Largo A. Gemelli 8, Rome, Italy
| | - Dimitar G. Efremov
- Department of Molecular Hematology, International Centre for Genetic Engineering & Biotechnology, Campus A. Buzzati-Traverso, Rome, Italy
| |
Collapse
|
37
|
Stephens DM, Byrd JC. Improving the Treatment Outcome of Patients with Chronic Lymphocytic Leukemia Through Targeted Antibody Therapy. Hematol Oncol Clin North Am 2013; 27:303-27. [DOI: 10.1016/j.hoc.2012.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
38
|
Seidel UJE, Schlegel P, Lang P. Natural killer cell mediated antibody-dependent cellular cytotoxicity in tumor immunotherapy with therapeutic antibodies. Front Immunol 2013; 4:76. [PMID: 23543707 PMCID: PMC3608903 DOI: 10.3389/fimmu.2013.00076] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 03/08/2013] [Indexed: 12/11/2022] Open
Abstract
In the last decade several therapeutic antibodies have been Federal Drug Administration (FDA) and European Medicines Agency (EMEA) approved. Although their mechanisms of action in vivo is not fully elucidated, antibody-dependent cellular cytotoxicity (ADCC) mediated by natural killer (NK) cells is presumed to be a key effector function. A substantial role of ADCC has been demonstrated in vitro and in mouse tumor models. However, a direct in vivo effect of ADCC in tumor reactivity in humans remains to be shown. Several studies revealed a predictive value of FcγRIIIa-V158F polymorphism in monoclonal antibody treatment, indicating a potential effect of ADCC on outcome for certain indications. Furthermore, the use of therapeutic antibodies after allogeneic hematopoietic stem cell transplantation is an interesting option. Studying the role of the FcγRIIIa-V158F polymorphism and the influence of Killer-cell Immunoglobuline-like Receptor (KIR) receptor ligand incompatibility on ADCC in this approach may contribute to future transplantation strategies. Despite the success of approved second-generation antibodies in the treatment of several malignancies, efforts are made to further augment ADCC in vivo by antibody engineering. Here, we review currently used therapeutic antibodies for which ADCC has been suggested as effector function.
Collapse
Affiliation(s)
- Ursula J E Seidel
- Department of General Paediatrics, Oncology/Haematology, University Children's Hospital Tübingen Tübingen, Germany
| | | | | |
Collapse
|
39
|
Rafiq S, Butchar JP, Cheney C, Mo X, Trotta R, Caligiuri M, Jarjoura D, Tridandapani S, Muthusamy N, Byrd JC. Comparative assessment of clinically utilized CD20-directed antibodies in chronic lymphocytic leukemia cells reveals divergent NK cell, monocyte, and macrophage properties. THE JOURNAL OF IMMUNOLOGY 2013; 190:2702-11. [PMID: 23418626 DOI: 10.4049/jimmunol.1202588] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD20 is a widely validated, B cell-specific target for therapy in B cell malignancies. Rituximab is an anti-CD20 Ab that prolongs survival of chronic lymphocytic leukemia (CLL) patients when combined with chemotherapy. Ofatumumab and GA101 (obinutuzumab) are CD20-directed Abs currently being developed as alternative agents to rituximab in CLL based upon different properties of enhanced direct cell death, NK cell-mediated Ab-dependent cellular cytotoxicity, or complement-dependent cytotoxicity. Despite widespread study, ofatumumab and GA101 have not been compared with each other, nor studied for their interactions with monocytes and macrophages which are critical for the efficacy of anti-CD20 Abs in murine models. In CLL cells, we show that direct cell death and complement-dependent cytotoxicity are greatest with GA101 and ofatumumab, respectively. GA101 promotes enhanced NK cell activation and Ab-dependent cellular cytotoxicity at high Ab concentrations. Ofatumumab elicits superior Ab-dependent cellular phagocytosis with monocyte-derived macrophages. GA101 demonstrated reduced activation of monocytes with diminished pERK, TNF-α release, and FcγRIIa recruitment to lipid rafts. These data demonstrate that GA101 and ofatumumab are both superior to rituximab against CLL cells via different mechanisms of potential tumor elimination. These findings bear relevance to potential combination strategies with each of these anti-CD20 Abs in the treatment of CLL.
Collapse
Affiliation(s)
- Sarwish Rafiq
- Integrated Biomedical Science Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Herishanu Y, Kay S, Dezorella N, Baron S, Hazan-Halevy I, Porat Z, Trestman S, Perry C, Braunstein R, Deutsch V, Polliack A, Naparstek E, Katz BZ. Divergence in CD19-Mediated Signaling Unfolds Intraclonal Diversity in Chronic Lymphocytic Leukemia, Which Correlates with Disease Progression. THE JOURNAL OF IMMUNOLOGY 2012; 190:784-93. [DOI: 10.4049/jimmunol.1200615] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
41
|
Human peripheral blood mononuclear cells exhibit heterogeneous CD52 expression levels and show differential sensitivity to alemtuzumab mediated cytolysis. PLoS One 2012; 7:e39416. [PMID: 22761788 PMCID: PMC3382607 DOI: 10.1371/journal.pone.0039416] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 05/24/2012] [Indexed: 11/25/2022] Open
Abstract
Alemtuzumab is a monoclonal antibody that targets cell surface CD52 and is effective in depleting lymphocytes by cytolytic effects in vivo. Although the cytolytic effects of alemtuzumab are dependent on the density of CD52 antigen on cells, there is scant information regarding the expression levels of CD52 on different cell types. In this study, CD52 expression was assessed on phenotypically distinct subsets of lymphoid and myeloid cells in peripheral blood mononuclear cells (PBMCs) from normal donors. Results demonstrate that subsets of PBMCs express differing levels of CD52. Quantitative analysis showed that memory B cells and myeloid dendritic cells (mDCs) display the highest number while natural killer (NK) cells, plasmacytoid dendritic cells (pDCs) and basophils have the lowest number of CD52 molecules per cell amongst lymphoid and myeloid cell populations respectively. Results of complement dependent cytolysis (CDC) studies indicated that alemtuzumab mediated profound cytolytic effects on B and T cells with minimal effect on NK cells, basophils and pDCs, correlating with the density of CD52 on these cells. Interestingly, despite high CD52 levels, mDCs and monocytes were less susceptible to alemtuzumab-mediated CDC indicating that antigen density alone does not define susceptibility. Additional studies indicated that higher expression levels of complement inhibitory proteins (CIPs) on these cells partially contributes to their resistance to alemtuzumab mediated CDC. These results indicate that alemtuzumab is most effective in depleting cells of the adaptive immune system while leaving innate immune cells relatively intact.
Collapse
|
42
|
Veliz M, Pinilla-Ibarz J. Treatment of relapsed or refractory chronic lymphocytic leukemia. Cancer Control 2012; 19:37-53. [PMID: 22143061 DOI: 10.1177/107327481201900105] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Recent advances in the treatment of chronic lymphocytic leukemia (CLL) have moved beyond the traditional use of alkylating agents and purine analogs into regimens combining these two chemotherapy classes with monoclonal antibodies. METHODS This article reviews treatments options for patients with relapsed or refractory CLL. RESULTS Several studies have investigated novel agents in treating patients with 17p deletion, TP53 mutation, and fludarabine-refractory CLL, as well as patients with suboptimal response to intense treatment. These investigational agents include rituximab, alemtuzumab, ofatumumab, bendamustine, high-dose methylprednisolone, lenalidomide, lumiliximab, cyclin-dependent kinase inhibitors, small modular immunopharmaceuticals, Bcl-2 inhibitors, and histone deacetylase inhibitors. While these newer drugs and combination therapies have shown promise as treatment options for CLL, additional studies are needed to determine the immunosuppression, toxicities, and infections associated with their use. CONCLUSIONS Despite improvement in initial overall response rates, most patients relapse and require further treatment. CLL remains incurable with standard therapies due to development of disease refractoriness. As such, novel approaches such as those noted above warrant continued research to improve outcomes for patients with CLL.
Collapse
Affiliation(s)
- Marays Veliz
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | | |
Collapse
|
43
|
Abstract
BACKGROUND Historically, alkylator-based therapy has been used to treat patients with chronic lymphocytic leukemia (CLL). More effective therapies, such as the use of monoclonal antibodies in combination with chemotherapy, have been shown to prolong both progression-free survival and overall survival. Improvements in the identification of prognostic markers for CLL, as well as novel combinations for chemoimmunotherapy regimens, have improved the outcome for patients with CLL. METHODS We examine the diagnosis of CLL, the role of prognostic factors in determining treatment goals, and current data on front-line management of CLL. RESULTS The benefits of single-agent and combination therapies are associated with prolonged progression-free and overall survival. While more aggressive management may therefore be warranted, each patient's comorbidities and performance status must be weighed against the benefits, availability, cost, treatment goals, and incidence of adverse effects associated with each therapy. CONCLUSIONS New single agents and novel treatment combinations have shown promising results in phase I/II studies. The ultimate therapeutic goals of prolonged survival and improved quality of life will be validated only by ongoing clinical and laboratory research and by continuous enrollment of patients in clinical trials.
Collapse
Affiliation(s)
- Sheetal Desai
- Department of Pharmacy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | | |
Collapse
|
44
|
Nguyen TH, Havari E, McLaren R, Zhang M, Jiang Y, Madden SL, Roberts B, Kaplan J, Shankara S. Alemtuzumab induction of intracellular signaling and apoptosis in malignant B lymphocytes. Leuk Lymphoma 2011; 53:699-709. [PMID: 21916527 DOI: 10.3109/10428194.2011.623253] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The molecular changes induced by alemtuzumab following binding of CD52 on B tumor cells were investigated. Alemtuzumab alone had no detectable impact on cell signaling but cross-linking of alemtuzumab on the surface of B tumor lines with anti-human Fc antibodies induced a transient Ca(2+) flux followed by phosphorylation of several kinases involved in stress and survival pathways, and expression of associated proteins including TNF-α. Cross-linking of alemtuzumab also induced capping and caspase-dependent apoptosis of the tumor lines. When using primary cells from B-CLL patients, alemtuzumab alone was capable of inducing protein phosphorylation and apoptosis through the cross-linking of alemtuzumab by FcγRIIb receptors on B-CLL cells. Apoptosis was prevented by blocking of FcγRIIb receptors with anti-CD32 antibody. Overall, our results indicate that cross-linking of alemtuzumab on B tumor cells can occur naturally through Fc receptor interaction and leads to the activation of specific cellular pathways and induction of apoptosis.
Collapse
|
45
|
Fernández-Marrero Y, López-Requena A. Lonely killers: effector cell- and complement-independent non-proapoptotic cytotoxic antibodies inducing membrane lesions. MAbs 2011; 3:528-34. [PMID: 22123064 DOI: 10.4161/mabs.3.6.17770] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The majority of the most effective monoclonal antibodies (mAbs) currently in the clinics bind to cancer or immune cells. Classic mechanisms of cell killing by therapeutic mAbs include antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity and induction of apoptosis by engagement of specific cell ligands. A few reports have described mAbs whose cytotoxic activity is Fc-independent and that do not induce the morphological and biochemical changes associated with the apoptosis-type of cell death. Even fewer works describe mAbs able to directly induce membrane lesions. Here, we discuss the available data on those molecules and their cell killing activity, with particular attention to the case of a mAb specific for the tumor-associated N-glycolyl (Neu5Gc)-GM3 ganglioside (GM3(Neu5Gc)). Some similarities are found in the cell death pathways triggered by these mAbs, but data are not abundant. We conclude that the usefulness of mAbs with a direct cytotoxic activity for immunotherapeutic strategies deserves deeper research.
Collapse
|
46
|
Abstract
Flow cytometry has had an impact upon all areas of clinical pathology and now, in the 21st century, it is truly coming of age. This study reviews the application of flow cytometry within clinical pathology with an emphasis upon haematology and immunology. The basic principles of flow cytometry are discussed, including the principles and considerations of the flow-cell and hydrodynamic focusing, detector layout and function, use of fluorochromes and multicolour flow cytometry (spectral overlap and colour compensation), alongside the strategies available for sample preparation, data acquisition and analysis, reporting of results, internal quality control, external quality assessment and flow sorting. The practice of flow cytometry is discussed, including the principles and pitfalls associated with leukocyte immunophenotyping for leukaemia and lymphoma diagnosis, immune deficiency, predicting and monitoring response to monoclonal antibody therapy, rare event detection and screening for genetic disease. Each section is illustrated with a case study. Future directions are also discussed.
Collapse
Affiliation(s)
- Paul F Virgo
- Department of Immunology and Immunogenetics, Southmead Hospital, Westbury-on-Trym, Bristol BS10 5NB
| | - Graham J Gibbs
- Department of Haematology, Musgrove Park Hospital, Taunton, Somerset TA1 5DA, UK
| |
Collapse
|
47
|
Daha NA, Banda NK, Roos A, Beurskens FJ, Bakker JM, Daha MR, Trouw LA. Complement activation by (auto-) antibodies. Mol Immunol 2011; 48:1656-65. [DOI: 10.1016/j.molimm.2011.04.024] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 04/15/2011] [Accepted: 04/20/2011] [Indexed: 12/24/2022]
|
48
|
Abstract
Cancer immunotherapy consists of approaches that modify the host immune system, and/or the utilization of components of the immune system, as cancer treatment. During the past 25 years, 17 immunologic products have received regulatory approval based on anticancer activity as single agents and/or in combination with chemotherapy. These include the nonspecific immune stimulants BCG and levamisole; the cytokines interferon-α and interleukin-2; the monoclonal antibodies rituximab, ofatumumab, alemtuzumab, trastuzumab, bevacizumab, cetuximab, and panitumumab; the radiolabeled antibodies Y-90 ibritumomab tiuxetan and I-131 tositumomab; the immunotoxins denileukin diftitox and gemtuzumab ozogamicin; nonmyeloablative allogeneic transplants with donor lymphocyte infusions; and the anti-prostate cancer cell-based therapy sipuleucel-T. All but two of these products are still regularly used to treat various B- and T-cell malignancies, and numerous solid tumors, including breast, lung, colorectal, prostate, melanoma, kidney, glioblastoma, bladder, and head and neck. Positive randomized trials have recently been reported for idiotype vaccines in lymphoma and a peptide vaccine in melanoma. The anti-CTLA-4 monoclonal antibody ipilumumab, which blocks regulatory T-cells, is expected to receive regulatory approval in the near future, based on a randomized trial in melanoma. As the fourth modality of cancer treatment, biotherapy/immunotherapy is an increasingly important component of the anticancer armamentarium.
Collapse
Affiliation(s)
- Robert O Dillman
- Hoag Cancer Institute of Hoag Hospital , Newport Beach, California 92658, USA.
| |
Collapse
|
49
|
Alinari L, Yu B, Christian BA, Yan F, Shin J, Lapalombella R, Hertlein E, Lustberg ME, Quinion C, Zhang X, Lozanski G, Muthusamy N, Prætorius-Ibba M, O'Connor OA, Goldenberg DM, Byrd JC, Blum KA, Baiocchi RA. Combination anti-CD74 (milatuzumab) and anti-CD20 (rituximab) monoclonal antibody therapy has in vitro and in vivo activity in mantle cell lymphoma. Blood 2011; 117:4530-41. [PMID: 21228331 PMCID: PMC3099572 DOI: 10.1182/blood-2010-08-303354] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2010] [Accepted: 12/18/2010] [Indexed: 02/07/2023] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell malignancy with a median survival of 3 years despite chemoimmunotherapy. Rituximab, a chimeric anti-CD20 monoclonal antibody (mAb), has shown only modest activity as single agent in MCL. The humanized mAb milatuzumab targets CD74, an integral membrane protein linked with promotion of B-cell growth and survival, and has shown preclinical activity against B-cell malignancies. Because rituximab and milatuzumab target distinct antigens and potentially signal through different pathways, we explored a preclinical combination strategy in MCL. Treatment of MCL cell lines and primary tumor cells with immobilized milatuzumab and rituximab resulted in rapid cell death, radical oxygen species generation, and loss of mitochondrial membrane potential. Cytoskeletal distrupting agents significantly reduced formation of CD20/CD74 aggregates, cell adhesion, and cell death, highlighting the importance of actin microfilaments in rituximab/milatuzumab-mediated cell death. Cell death was independent of caspase activation, Bcl-2 family proteins or modulation of autophagy. Maximal inhibition of p65 nuclear translocation was observed with combination treatment, indicating disruption of the NF-κB pathway. Significant in vivo therapeutic activity of combination rituximab and milatuzumab was demonstrated in a preclinical model of MCL. These data support clinical evaluation of combination milatuzumab and rituximab therapy in MCL.
Collapse
MESH Headings
- Antibodies, Immobilized/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Antibodies, Monoclonal, Murine-Derived/pharmacology
- Antigens, CD20/immunology
- Antigens, CD20/metabolism
- Antigens, Differentiation, B-Lymphocyte/immunology
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Antineoplastic Agents/pharmacology
- Cell Death/drug effects
- Cell Death/immunology
- Cell Line, Tumor
- Cytoskeleton/drug effects
- Cytoskeleton/immunology
- Cytoskeleton/metabolism
- Drug Therapy, Combination
- Flow Cytometry
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Humans
- In Vitro Techniques
- Lymphoma, Mantle-Cell/drug therapy
- Lymphoma, Mantle-Cell/pathology
- Membrane Potential, Mitochondrial/drug effects
- Membrane Potential, Mitochondrial/immunology
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/immunology
- Reactive Oxygen Species/metabolism
- Rituximab
Collapse
Affiliation(s)
- Lapo Alinari
- Division of Hematology, Department of Medicine, College of Medicine, The Ohio State University, Columbus, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Denkert C, Darb-Esfahani S, Loibl S, Anagnostopoulos I, Jöhrens K. Anti-cancer immune response mechanisms in neoadjuvant and targeted therapy. Semin Immunopathol 2011; 33:341-51. [PMID: 21499853 DOI: 10.1007/s00281-011-0261-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 03/07/2011] [Indexed: 02/04/2023]
Abstract
Several studies suggest that the progression of malignant tumors as well as the response to chemotherapy and targeted therapy is critically dependent on the immunological parameters that are derived from the host immune system as well as a modulation of the immune system by therapeutic antibodies. It has been shown for many tumor types that the presence of a lymphocytic infiltrate in different types of cancers is a positive factor for clinical outcome and that the response to neoadjuvant chemotherapy is increased in a tumor with a prominent pretherapeutic infiltrate. Furthermore, new targeted therapies in breast cancer, such as trastuzumab, as well as in hematological malignancies, such as rituximab and alemtuzumab, have been shown to interact with immunological pathways, and this interaction is critical for response and clinical outcome. In neoplasms of lymphoid and hematopoietic tissues, targeted therapies not only reduce toxic effects on normal tissues but also lead to modulations of the immune system depending on the target molecule, its physiological function and cellular distribution. This review gives an overview on clinical data on response to classical chemotherapy as well as molecular targeted therapy and its interaction with the immune system.
Collapse
Affiliation(s)
- Carsten Denkert
- Institute of Pathology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | | | | | | | | |
Collapse
|