1
|
Anderson GSF, Chapman MA. T cell-redirecting therapies in hematological malignancies: Current developments and novel strategies for improved targeting. Mol Ther 2024; 32:2856-2891. [PMID: 39095991 PMCID: PMC11403239 DOI: 10.1016/j.ymthe.2024.07.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
T cell-redirecting therapies (TCRTs), such as chimeric antigen receptor (CAR) or T cell receptor (TCR) T cells and T cell engagers, have emerged as a highly effective treatment modality, particularly in the B and plasma cell-malignancy setting. However, many patients fail to achieve deep and durable responses; while the lack of truly unique tumor antigens, and concurrent on-target/off-tumor toxicities, have hindered the development of TCRTs for many other cancers. In this review, we discuss the recent developments in TCRT targets for hematological malignancies, as well as novel targeting strategies that aim to address these, and other, challenges.
Collapse
Affiliation(s)
| | - Michael A Chapman
- MRC Toxicology Unit, University of Cambridge, Cambridge CB2 1QR, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK; Addenbrooke's Hospital, Cambridge Universities Foundation Trust, Cambridge CB2 0QQ, UK.
| |
Collapse
|
2
|
Tian XP, Cao Y, Cai J, Zhang YC, Zou QH, Wang JN, Fang Y, Wang JH, Guo SB, Cai QQ. Novel target and treatment agents for natural killer/T-cell lymphoma. J Hematol Oncol 2023; 16:78. [PMID: 37480137 PMCID: PMC10362755 DOI: 10.1186/s13045-023-01483-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/19/2023] [Indexed: 07/23/2023] Open
Abstract
The rapidly increasing use of high-throughput screening had produced a plethora of expanding knowledge on the molecular basis of natural killer/T-cell lymphoma (NKTCL), which in turn has revolutionized the treatment. Specifically, the use of asparaginase-containing regimens has led to substantial improvement in survival outcomes in NKTCL patients. Novel treatment strategies that are currently under development include cell-surface-targeted antibodies, immune checkpoint inhibitors, Epstein-Barr virus targeted cytotoxic T lymphocyte, immunomodulatory agents, chimeric antigen receptor T cells, signaling pathway inhibitors and epigenetic targeted agents. In almost all cases, initial clinical studies of newly developed treatment are conducted in patients relapsed, and refractory NKTCL due to very limited treatment options. This review summarizes the results of these novel treatments for NKTCL and discusses their potential for likely use in NKTCL in a wider setting in the future.
Collapse
Affiliation(s)
- Xiao-Peng Tian
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yi Cao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jun Cai
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yu-Chen Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Qi-Hua Zou
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jin-Ni Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yu Fang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jia-Hui Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Song-Bin Guo
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Qing-Qing Cai
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.
| |
Collapse
|
3
|
Ma XC, Lv X, Li Y. Development of CD30 CAR-T cells in refractory or relapsed Hodgkin's lymphoma. Expert Rev Hematol 2023; 16:1017-1023. [PMID: 37888882 DOI: 10.1080/17474086.2023.2276210] [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: 06/21/2023] [Accepted: 10/24/2023] [Indexed: 10/28/2023]
Abstract
INTRODUCTION After therapy, approximately 15% of individuals with Hodgkin's lymphoma (HL) develop relapsed or drug-resistant Hodgkin's lymphoma (r/rHL). r/rHL has a high fatality rate and poor therapeutic prognosis. CD30 CAR-T-cell therapy has emerged as a new way to treat r/rHL in recent years. However, CD30CAR-T cells are still being explored in clinical trials. To help more patients, this review focuses on current CD30CAR-T-cell advancements as well as clinical breakthroughs in treatment of r/rHL. AREAS COVERED This research examines the mechanism of action of CD30 CAR-T cells, their function in the real-world therapy of r/rHL, and the influence of different treatment regimens on treatment results. EXPERT OPINION There has been much research into CD30 CAR-T cells as a result of their successful use in treatment of r/rHL. This research has helped us to understand CD30 CAR-T-cell safety as well as the management options available before and after its administration to increase patient survival and reduce side effects.
Collapse
Affiliation(s)
- Xiao Chen Ma
- Department of Haematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Xiao Lv
- Department of Haematology, Shan dong Provincial Hospital Affiliated to Shan dong First Medical University; Shan dong Provincial Hospital, Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Ying Li
- Department of Haematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| |
Collapse
|
4
|
Guan X, Hu H, Tian M, Zhuang H, Ding C, Yu S. Differentially expressed long noncoding RNAs in RAW264.7 macrophages during Brucella infection and functional analysis on the bacterial intracellular replication. Sci Rep 2022; 12:21320. [PMID: 36494502 PMCID: PMC9734652 DOI: 10.1038/s41598-022-25932-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are a group of functional RNA molecules without protein-coding potential and play vital roles in majority of biological processes. To date, the expression profiles of lncRNAs and their influence on Brucella replication in RAW264.7 cells are poorly understood. In this study, we performed high-throughput transcriptome analysis to investigate the differentially expressed lncRNAs associated with Brucella abortus S2308 infection. Of these, 8, 6, 130 and 94 cellular lncRNAs were differentially expressed at 4, 8, 24 and 48 h post-infection, respectively. Moreover, 1918 protein-coding genes are predicted as potential cis target genes of differentially expressed lncRNAs by searching protein-coding genes located at upstream and downstream of lncRNA loci on the chromosome DNA of Mus musculus. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses indicated that majority of lncRNA target genes were associated with B. abortus infection. Fourteen lncRNAs from transcriptome data were selected for qRT-PCR verification, confirming 13 were differentially expressed. Animal experiments revealed three were differentially expressed in vivo by qRT-PCR analysis. Furthermore, knockdown of LNC_000428 by CRISPR/dCas9 inhibition or Locked Nucleic Acids transfection downregulated Tnfrsf8 expression at mRNA level and increased Brucella intracellular replication. Thus, we provide a novel evidence that lncRNAs induced by Brucella-infection function on Brucella intracellular replication.
Collapse
Affiliation(s)
- Xiang Guan
- grid.410727.70000 0001 0526 1937Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Minhang District, Shanghai, 200241 China
| | - Hai Hu
- grid.410727.70000 0001 0526 1937Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Minhang District, Shanghai, 200241 China
| | - Minxing Tian
- grid.410727.70000 0001 0526 1937Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Minhang District, Shanghai, 200241 China
| | - Hongxu Zhuang
- grid.410727.70000 0001 0526 1937Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Minhang District, Shanghai, 200241 China
| | - Chan Ding
- grid.410727.70000 0001 0526 1937Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Minhang District, Shanghai, 200241 China
| | - Shengqing Yu
- grid.410727.70000 0001 0526 1937Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Minhang District, Shanghai, 200241 China
| |
Collapse
|
5
|
Wang L, Zhao J, Schank M, Khanal S, Dang X, Cao D, Nguyen LNT, Zhang Y, Wu XY, Adkins JL, Brueggeman J, Zhang J, Ning S, El Gazzar M, Moorman JP, Yao ZQ. Identification of virus-specific B-cell epitopes by convalescent plasma from COVID-19 patients. Mol Immunol 2022; 152:215-223. [PMID: 36379129 PMCID: PMC9630139 DOI: 10.1016/j.molimm.2022.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/11/2022] [Accepted: 10/30/2022] [Indexed: 11/05/2022]
Abstract
Identification of immunologic epitopes against SARS-CoV-2 is crucial for the discovery of diagnostic, therapeutic, and preventive targets. In this study, we used a pan-coronavirus peptide microarray to screen for potential B-cell epitopes and validated the results with peptide-based ELISA. Specifically, we identified three linear B-cell epitopes on the SARS-CoV-2 proteome, which were recognized by convalescent plasma from COVID-19 patients. Interestingly, two epitopes (S 809-823 and R1ab 909-923) strongly reacted to convalescent plasma collected at the early phase (< 90 days) of COVID-19 symptom onset, whereas one epitope (M 5-19) reacted to convalescent plasma collected > 90 days after COVID-19 symptom onset. Neutralization assays using antibody depletion with the identified spike (S) peptides revealed that three S epitopes (S 557-571, S 789-803, and S 809-823) elicited neutralizing antibodies in COVID-19 patients. However, the levels of virus-specific antibody targeting S 789-803 only positively correlated with the neutralizing rates at the early phase (<60 days) after disease onset, and the antibody titers diminished quickly with no correlation to the neutralizing activity beyond two months after recovery from COVID-19. Importantly, stimulation of peripheral blood mononuclear cells from COVID-19-recovered patients with these SARS-CoV-2 S peptides resulted in poor virus-specific B cell activation, proliferation, differentiation into memory B cells, and production of immunoglobulin G (IgG) antibodies, despite the B-cells being functionally competent as demonstrated by their response to non-specific stimulation. Taken together, these findings indicate that these newly identified SARS-CoV-2-specific B-cell epitopes can elicit neutralizing antibodies, with titers and/or neutralizing activities declining significantly within 2-3 months in the convalescent plasma of COVID-19 patients.
Collapse
Affiliation(s)
- Ling Wang
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA.
| | - Juan Zhao
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Madison Schank
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Sushant Khanal
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Xindi Dang
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Dechao Cao
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Lam N T Nguyen
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Yi Zhang
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Xiao Y Wu
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - James L Adkins
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Justin Brueggeman
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Jinyu Zhang
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Shunbin Ning
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Mohamed El Gazzar
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Jonathan P Moorman
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA; Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN 37614, USA
| | - Zhi Q Yao
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA; Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN 37614, USA.
| |
Collapse
|
6
|
Filling the Gap: The Immune Therapeutic Armamentarium for Relapsed/Refractory Hodgkin Lymphoma. J Clin Med 2022; 11:jcm11216574. [PMID: 36362802 PMCID: PMC9656939 DOI: 10.3390/jcm11216574] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/30/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Despite years of clinical progress which made Hodgkin lymphoma (HL) one of the most curable malignancies with conventional chemotherapy, refractoriness and recurrence may still affect up to 20–30% of patients. The revolution brought by the advent of immunotherapy in all kinds of neoplastic disorders is more than evident in this disease because anti-CD30 antibodies and checkpoint inhibitors have been able to rescue patients previously remaining without therapeutic options. Autologous hematopoietic cell transplantation still represents a significant step in the treatment algorithm for chemosensitive HL; however, the possibility to induce complete responses after allogeneic transplant procedures in patients receiving reduced-intensity conditioning regimens informs on its sensitivity to immunological control. Furthermore, the investigational application of adoptive T cell transfer therapies paves the way for future indications in this setting. Here, we seek to provide a fresh and up-to-date overview of the new immunotherapeutic agents dominating the scene of relapsed/refractory HL. In this optic, we will also review all the potential molecular mechanisms of tumor resistance, theoretically responsible for treatment failures, and we will discuss the place of allogeneic stem cell transplantation in the era of novel therapies.
Collapse
|
7
|
Mitteldorf C, Kampa F, Ströbel P, Schön MP, Kempf W. Intraindividual variability of
CD30
expression in mycosis fungoides –implications for diagnostic evaluation and therapy. Histopathology 2022; 81:55-64. [DOI: 10.1111/his.14660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/24/2022] [Accepted: 04/04/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Christina Mitteldorf
- Department of Dermatology, Venereology and Allergology University Medical Center Göttingen Germany
| | - Franziska Kampa
- Department of Dermatology, Venereology and Allergology University Medical Center Göttingen Germany
| | | | - Michael P. Schön
- Department of Dermatology, Venereology and Allergology University Medical Center Göttingen Germany
- Lower Saxony Institute of Occupational Dermatology
| | - Werner Kempf
- Kempf und Pfaltz Histologische Diagnostik, Affolternstrasse 56, CH‐8050 Zürich Switzerland
- Department of Dermatology University Hospital Zurich CH‐8091 Zurich Switzerland
| |
Collapse
|
8
|
Shademan B, Karamad V, Nourazarian A, Avcı CB. CAR T Cells: Cancer Cell Surface Receptors Are the Target for Cancer Therapy. Adv Pharm Bull 2021; 12:476-489. [PMID: 35935042 PMCID: PMC9348524 DOI: 10.34172/apb.2022.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/12/2021] [Accepted: 08/17/2021] [Indexed: 11/09/2022] Open
Abstract
Immunotherapy has become a prominent strategy for the treatment of cancer. A method that improves the immune system's ability to attack a tumor (Enhances antigen binding). Targeted killing of malignant cells by adoptive transfer of chimeric antigen receptor (CAR) T cells is a promising immunotherapy technique in the treatment of cancers. For this purpose, the patient's immune cells, with genetic engineering aid, are loaded with chimeric receptors that have particular antigen binding and activate cytotoxic T lymphocytes. That increases the effectiveness of immune cells and destroying cancer cells. This review discusses the basic structure and function of CAR-T cells and how antigenic targets are identified to treat different cancers and address the disadvantages of this treatment for cancer.
Collapse
Affiliation(s)
- Behrouz Shademan
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Vahidreza Karamad
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Alireza Nourazarian
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Cigir Biray Avcı
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| |
Collapse
|
9
|
Belmonte B, Cancila V, Gulino A, Navari M, Arancio W, Macor P, Balduit A, Capolla S, Morello G, Vacca D, Ferrara I, Bertolazzi G, Balistreri CR, Amico P, Ferrante F, Maiorana A, Salviato T, Piccaluga PP, Mangogna A. Constitutive PSGL-1 Correlates with CD30 and TCR Pathways and Represents a Potential Target for Immunotherapy in Anaplastic Large T-Cell Lymphoma. Cancers (Basel) 2021; 13:2958. [PMID: 34204843 PMCID: PMC8231564 DOI: 10.3390/cancers13122958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/13/2022] Open
Abstract
Due to the high expression of P-selectin glycoprotein ligand-1 (PSGL-1) in lymphoproliferative disorders and in multiple myeloma, it has been considered as a potential target for humoral immunotherapy, as well as an immune checkpoint inhibitor in T-cells. By investigating the expression of SELPLG in 678 T- and B-cell samples by gene expression profiling (GEP), further supported by tissue microarray and immunohistochemical analysis, we identified anaplastic large T-cell lymphoma (ALCL) as constitutively expressing SELPLG at high levels. Moreover, GEP analysis in CD30+ ALCLs highlighted a positive correlation of SELPLG with TNFRSF8 (CD30-coding gene) and T-cell receptor (TCR)-signaling genes (LCK, LAT, SYK and JUN), suggesting that the common dysregulation of TCR expression in ALCLs may be bypassed by the involvement of PSGL-1 in T-cell activation and survival. Finally, we evaluated the effects elicited by in vitro treatment with two anti-PSGL-1 antibodies (KPL-1 and TB5) on the activation of the complement system and induction of apoptosis in human ALCL cell lines. In conclusion, our data demonstrated that PSGL-1 is specifically enriched in ALCLs, altering cell motility and viability due to its involvement in CD30 and TCR signaling, and it might be considered as a promising candidate for novel immunotherapeutic approaches in ALCLs.
Collapse
Affiliation(s)
- Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy; (B.B.); (V.C.); (A.G.); (G.M.); (D.V.); (I.F.); (G.B.); (F.F.)
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy; (B.B.); (V.C.); (A.G.); (G.M.); (D.V.); (I.F.); (G.B.); (F.F.)
| | - Alessandro Gulino
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy; (B.B.); (V.C.); (A.G.); (G.M.); (D.V.); (I.F.); (G.B.); (F.F.)
| | - Mohsen Navari
- Department of Medical Biotechnology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh 95196 33787, Iran;
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh 95196 33787, Iran
- Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad 91766 99199, Iran
| | - Walter Arancio
- Advanced Data Analysis Group, Fondazione Ri.MED, 90133 Palermo, Italy;
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (P.M.); (A.B.); (S.C.)
| | - Andrea Balduit
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (P.M.); (A.B.); (S.C.)
| | - Sara Capolla
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (P.M.); (A.B.); (S.C.)
| | - Gaia Morello
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy; (B.B.); (V.C.); (A.G.); (G.M.); (D.V.); (I.F.); (G.B.); (F.F.)
| | - Davide Vacca
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy; (B.B.); (V.C.); (A.G.); (G.M.); (D.V.); (I.F.); (G.B.); (F.F.)
| | - Ines Ferrara
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy; (B.B.); (V.C.); (A.G.); (G.M.); (D.V.); (I.F.); (G.B.); (F.F.)
| | - Giorgio Bertolazzi
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy; (B.B.); (V.C.); (A.G.); (G.M.); (D.V.); (I.F.); (G.B.); (F.F.)
| | - Carmela Rita Balistreri
- Department of BioMedicine, Neuroscience, and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo, Italy;
| | - Paolo Amico
- Department of Pathology, Cannizzaro Hospital, 95126 Catania, Italy;
| | - Federica Ferrante
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy; (B.B.); (V.C.); (A.G.); (G.M.); (D.V.); (I.F.); (G.B.); (F.F.)
| | - Antonino Maiorana
- Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena and Reggio Emilia, 41121 Modena, Italy; (A.M.); (T.S.)
| | - Tiziana Salviato
- Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena and Reggio Emilia, 41121 Modena, Italy; (A.M.); (T.S.)
| | - Pier Paolo Piccaluga
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, 40126 Bologna, Italy;
- Section of Genomics and Personalized Medicine, Istituto Euro-Mediterraneo di Scienza e Tecnologia (IEMEST), 90139 Palermo, Italy
- Department of Pathology, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, 00622 Juja, Kenya
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) “Burlo Garofolo”, 34137 Trieste, Italy
| |
Collapse
|
10
|
EBF1 drives hallmark B cell gene expression by enabling the interaction of PAX5 with the MLL H3K4 methyltransferase complex. Sci Rep 2021; 11:1537. [PMID: 33452395 PMCID: PMC7810865 DOI: 10.1038/s41598-021-81000-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/28/2020] [Indexed: 12/15/2022] Open
Abstract
PAX5 and EBF1 work synergistically to regulate genes that are involved in B lymphocyte differentiation. We used the KIS-1 diffuse large B cell lymphoma cell line, which is reported to have elevated levels of PAX5 expression, to investigate the mechanism of EBF1- and PAX5-regulated gene expression. We demonstrate the lack of expression of hallmark B cell genes, including CD19, CD79b, and EBF1, in the KIS-1 cell line. Upon restoration of EBF1 expression we observed activation of CD19, CD79b and other genes with critical roles in B cell differentiation. Mass spectrometry analyses of proteins co-immunoprecipitated with PAX5 in KIS-1 identified components of the MLL H3K4 methylation complex, which drives histone modifications associated with transcription activation. Immunoblotting showed a stronger association of this complex with PAX5 in the presence of EBF1. Silencing of KMT2A, the catalytic component of MLL, repressed the ability of exogenous EBF1 to activate transcription of both CD19 and CD79b in KIS-1 cells. We also find association of PAX5 with the MLL complex and decreased CD19 expression following silencing of KMT2A in other human B cell lines. These data support an important role for the MLL complex in PAX5-mediated transcription regulation.
Collapse
|
11
|
Gong J, Guo F, Cheng W, Fan H, Miao Q, Yang J. Preliminary biological evaluation of 123I-labelled anti-CD30-LDM in CD30-positive lymphomas murine models. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2020; 48:408-414. [PMID: 31913714 DOI: 10.1080/21691401.2019.1709857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/19/2019] [Indexed: 01/21/2023]
Abstract
Overexpression of CD30 has been reported on the surface of some T-cell lymphomas, especially on Hodgkin's lymphoma (HL) and anaplastic large cell lymphoma (ALCL). CD30 targeted immunotherapy has good clinical therapy response. We have produced a novel antibody drug conjugates (ADCs)-anti-CD30-LDM, which shows attractive tumour-targeting capability and extremely potent antitumor efficacy. To further investigate biological characteristics and promote clinical translation of anti-CD30-LDM, we constructed a radiolabeled 123I-anti-CD30-LDM to evaluate the biodistribution characteristics. The anti-CD30-LDM was radioiodinated by the Iodogen method. The radiochemical purity of 123I-anti-CD30-LDM was more over 98%, and the specific activity of 240.5 MBq/mg. The stability and the specificity of 123I-anti-CD30-LDM were evaluated in vitro. Cellular binding assays were used to evaluate the binding capabilities in CD30-positive Karpas299 cells and CD30-negative Raji cells. B-NDG mice bearing Karpas 299 and Raji xenografts were used for in vivo biodistribution studies. Our results demonstrated that anti-CD30-LDM as an ideal ADC targeted to CD30, which was labelled easily with 123I and obtained the sufficient yields. The 123I-anti-CD30-LDM preserved specific binding to CD30 in vitro and uptake in tumour xenografts in B-NDG mice. These results are encouraging for anti-CD30-LDM as a promising clinical translational candidate for various CD30 positive lymphomas and other diseases.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/chemistry
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/pharmacology
- Humans
- Iodine Radioisotopes/chemistry
- Iodine Radioisotopes/pharmacokinetics
- Iodine Radioisotopes/pharmacology
- Ki-1 Antigen/antagonists & inhibitors
- Lymphoma, Large-Cell, Anaplastic/drug therapy
- Lymphoma, Large-Cell, Anaplastic/metabolism
- Lymphoma, Large-Cell, Anaplastic/pathology
- Mice
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Radiopharmaceuticals/chemistry
- Radiopharmaceuticals/pharmacokinetics
- Radiopharmaceuticals/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jianhua Gong
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feihu Guo
- High Tech of Atom Co. Ltd, Beijing, China
| | | | | | - Qingfang Miao
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jigang Yang
- Nuclear Medicine Department, Beijing Friendship Hospital, Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Status of humoral and cellular immune markers in human T-cell lymphotropic virus type 1 (HTLV-1) asymptomatic carriers in northeastern Iran, Mashhad. J Neurovirol 2020; 26:863-869. [PMID: 33025348 DOI: 10.1007/s13365-020-00910-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 01/26/2020] [Accepted: 09/14/2020] [Indexed: 10/23/2022]
Abstract
It is estimated that about 10-20 million peoples are infected with human T-cell leukemia virus type 1 (HTLV-1) around the world and suffered from HTLV-related diseases. The present study was aimed to evaluate the cellular immunity, T-cell activation, humoral immunity, and inflammatory response hallmarks which affect HTLV-1-associated disease progression. A total of 78 participants were included in the study, comprising 39 HTLV-1 asymptomatic careers (ACs) and 39 healthy controls. The HTLV-proviral load (PVL) was determined via real-time PCR technique, and anti-HTLV antibody, sIL2R, sCD30, Neoptrin, hs-CRP, IgE, anti-VCA, anti-EBNA, and anti-EA were assessed by ELISA method. Mean PVL in ACs was 352.7 ± 418.7 copies/104 PBMCs. A significant higher level of sIL-2R was observed in ACs (P < 0.0001). Anti-VCA antibody titer in ACs and healthy controls was 80.72 ± 105.95 and 156.05 ± 130.71, respectively (P = 0.007). Intriguingly, suppression in ACs immune response was not observed. Resultantly, HTLV-1 infection has no effect on the humoral immune response in ACs but greater T-cell activation and function cellular responses were detected. Finally, more studies on various immune markers in adult T-cell leukemia/lymphoma (ATLL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) patients are greatly needed to illuminate the association of ACs' immune status with the development of the related diseases.
Collapse
|
13
|
Marchesini G, Nadali G, Facchinelli D, Candoni A, Cattaneo C, Cuccaro A, Fanci R, Farina F, Lessi F, Visentin A, Marchesi F, Prezioso L, Spolzino A, Tisi MC, Trastulli F, Verga L, Dargenio M, Busca A, Pagano L. Infections in patients with lymphoproliferative diseases treated with brentuximab vedotin: SEIFEM multicentric retrospective study. Leuk Lymphoma 2020; 61:3002-3005. [PMID: 32611212 DOI: 10.1080/10428194.2020.1786562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Gessica Marchesini
- Haematology Unit, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Gianpaolo Nadali
- Haematology Unit, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Davide Facchinelli
- Haematology Unit, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Anna Candoni
- Division of Haematology and Stem Cell Transplantation, University Hospital of Udine, Udine, Italy
| | | | - Annarosa Cuccaro
- Institute of Haematology, Fondazione Policlinico A. Gemelli - IRCCS-Università Cattolica S. Cuore, Roma, Italy
| | - Rosa Fanci
- Haematology Unit, Careggi Hospital and University of Firenze, Florence, Italy
| | - Francesca Farina
- Division of Haematology and Stem Cell Transplantation, IRCCS San Raffaele, Milano, Italy
| | - Federica Lessi
- Division of Haematology and Clinical Immunology, University of Padova, Padova, Italy
| | - Andrea Visentin
- Division of Haematology and Clinical Immunology, University of Padova, Padova, Italy
| | - Francesco Marchesi
- Haematology and Stem Cell Transplantation Unit, Regina Elena National Cancer Institute, Roma, Italy
| | - Lucia Prezioso
- Haematology and BMT Unit, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Angelica Spolzino
- Haematology and BMT Unit, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | | | - Fabio Trastulli
- Department of Clinical Medicine and Surgery, Federico II University Medical School, Napoli, Italy
| | - Luisa Verga
- Haematology and CTA ASST Monza, Milano Bicocca University, Ospedale San Gerardo, Monza, Italy
| | - Michela Dargenio
- Haematology and Stem Cell Transplantation Unit, 'Vito Fazzi' Hospital, Lecce, Italy
| | - Alessandro Busca
- SSD Division of Haematology and Stem Cell Transplantation, A.O.U. Città della Salute, Torino, Italy
| | - Livio Pagano
- Institute of Haematology, Fondazione Policlinico A. Gemelli - IRCCS-Università Cattolica S. Cuore, Roma, Italy
| | | |
Collapse
|
14
|
Hollander P, Ginman B, Molin D, Enblad G, Amini RM, Glimelius I. Precursor cells and implications of a T-cell inflamed immune response in the pre-malignant setting in Hodgkin lymphoma. Immunobiology 2020; 225:151872. [DOI: 10.1016/j.imbio.2019.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 01/08/2023]
|
15
|
Fu M, Tang L. Chimeric Antigen Receptor T Cell Immunotherapy for Tumor: A Review of Patent Literatures. Recent Pat Anticancer Drug Discov 2019; 14:60-69. [PMID: 30636615 DOI: 10.2174/1574892814666190111120908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/13/2018] [Accepted: 01/02/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Chimeric Antigen Receptor (CAR) T cell immunotherapy, as an innovative
method for tumor immunotherapy, acquires unprecedented clinical outcomes. Genetic modification not
only provides T cells with the antigen-binding function but also endows T cells with better
immunological functions both in solid and hematological cancer. However, the CAR T cell therapy is
not perfect because of several reasons, such as tumor immune microenvironment, and autologous
limiting factors of CAR T cells. Moreover, the safety of CAR T cells should be improved. OBJECTIVE Recently many patents and publications have reported the importance of CAR T cell immunotherapy. Based on the patents about CAR T cell immunotherapy, we conclude some methods for
designing the CAR which can provide useful information to readers. METHODS This review presents recent patents and publications, summarizes some specific antigens for
oncotherapy from patents and enumerate some approaches to treatment of immunosuppression and
reinforcing the immune response of CAR T cells. We also sum up some strategies for improving the
safety of CAR T cell immunotherapy. RESULTS CAR T cell immunotherapy as a neotype cellular immunotherapy has been proved effective in
oncotherapy and authorized by the FDA. Improvements in CAR designing have enhanced the
functions of CAR T cells. CONCLUSION This review, summarizing antigens and approaches to overcome defects of CAR T cell
immunotherapy from patents and publications, might contribute to a broad readership.
Collapse
Affiliation(s)
- Manxue Fu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, P.R. China. Address: No.174 Shazhengjie, Shapingba, Chongqing, China
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, P.R. China. Address: No.174 Shazhengjie, Shapingba, Chongqing, China
| |
Collapse
|
16
|
Hombach AA, Rappl G, Abken H. Blocking CD30 on T Cells by a Dual Specific CAR for CD30 and Colon Cancer Antigens Improves the CAR T Cell Response against CD30 - Tumors. Mol Ther 2019; 27:1825-1835. [PMID: 31331813 DOI: 10.1016/j.ymthe.2019.06.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 06/05/2019] [Accepted: 06/11/2019] [Indexed: 12/26/2022] Open
Abstract
Chimeric antigen receptor (CAR)-engineered T cells are efficacious in controlling advanced leukemia and lymphoma, however, they fail in the treatment of solid cancer, which is thought to be due to insufficient T cell activation. We revealed that the immune response of CAR T cells with specificity for carcinoembryonic antigen (CEA) was more efficacious against CEA+ cancer cells when simultaneously incubated with an anti-CD30 immunotoxin or anti-CD30 CAR T cells, although the targeted cancer cells lack CD30. The same effect was achieved when the anti-CD30 single-chain variable fragment (scFv) was integrated into the extracellular domain of the anti-CEA CAR. Improvement in T cell activation was due to interfering with the T cell CD30-CD30L interaction by the antagonistic anti-CD30 scFv HRS3; an agonistic anti-CD30 scFv or targeting the high-affinity interleukin-2 (IL-2) receptor was not effective. T cells with the anti-CD30/CEA CAR showed superior immunity against established CEA+ CD30- tumors in a mouse model. The concept is broadly applicable since anti-CD30/TAG72 CAR T cells also showed improved elimination of TAG72+ CD30- cancer cells. Taken together, targeting CD30 on CAR T cells by the HRS3 scFv within the anti-tumor CAR improves the redirected immune response against solid tumors.
Collapse
Affiliation(s)
- Andreas A Hombach
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.
| | - Gunter Rappl
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Hinrich Abken
- RCI, Regensburg Center for Interventional Immunology, University of Regensburg and University Hospital of Regensburg, Regensburg, Germany
| |
Collapse
|
17
|
Wang R, Li L, Duan A, Li Y, Liu X, Miao Q, Gong J, Zhen Y. Crizotinib enhances anti-CD30-LDM induced antitumor efficacy in NPM-ALK positive anaplastic large cell lymphoma. Cancer Lett 2019; 448:84-93. [PMID: 30742941 DOI: 10.1016/j.canlet.2019.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/28/2019] [Accepted: 02/01/2019] [Indexed: 12/21/2022]
Abstract
Combining antibody-drug conjugates (ADCs) with targeted small-molecule inhibitors can enhance antitumor effects beyond those attainable with monotherapy. In this study, we investigated the therapeutic combination of a CD30-targeting ADC (anti-CD30-lidamycin [LDM]) with a small-molecule inhibitor (crizotinib) of nucleophosmin-anaplastic lymphoma kinase NPM-ALK in CD30+/ALK+ anaplastic large cell lymphoma (ALCL). In vitro, anti-CD30-LDM showed strong synergistic antiproliferative activity when combined with crizotinib. Furthermore, treatment with anti-CD30-LDM plus crizotinib resulted in a stronger induction of cell apoptosis than monotherapy with either treatment. Western blot analysis revealed that ERK1/2 phosphorylation was increased in response to anti-CD30-LDM-induced DNA damage. Interestingly, the addition of crizotinib inhibited the expression of phosphorylated ERK1/2 and further augmented anti-CD30-LDM-mediated apoptosis, providing a potential synergistic mechanism for DNA-damaging agents combined with NPM-ALK inhibitors. In Karpas299 and SU-DHL-1 xenograft models, anti-CD30-LDM plus crizotinib was more effective in inhibiting tumor growth than either treatment alone. This research demonstrated for the first time that the combination of anti-CD30-LDM and crizotinib exhibits a synergistic inhibitory effect in tumor cells. These results provide scientific support for future clinical evaluations of anti-CD30-LDM, or other DNA-damaging agents, combined with NPM-ALK inhibitors.
Collapse
Affiliation(s)
- Rong Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liang Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Aijun Duan
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yi Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiujun Liu
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qingfang Miao
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Jianhua Gong
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Yongsu Zhen
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Maschmeyer G, De Greef J, Mellinghoff SC, Nosari A, Thiebaut-Bertrand A, Bergeron A, Franquet T, Blijlevens NMA, Maertens JA. Infections associated with immunotherapeutic and molecular targeted agents in hematology and oncology. A position paper by the European Conference on Infections in Leukemia (ECIL). Leukemia 2019; 33:844-862. [PMID: 30700842 PMCID: PMC6484704 DOI: 10.1038/s41375-019-0388-x] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/31/2018] [Accepted: 01/11/2019] [Indexed: 02/08/2023]
Abstract
A multitude of new agents for the treatment of hematologic malignancies has been introduced over the past decade. Hematologists, infectious disease specialists, stem cell transplant experts, pulmonologists and radiologists have met within the framework of the European Conference on Infections in Leukemia (ECIL) to provide a critical state-of-the-art on infectious complications associated with immunotherapeutic and molecular targeted agents used in clinical routine. For brentuximab vedotin, blinatumomab, CTLA4- and PD-1/PD-L1-inhibitors as well as for ibrutinib, idelalisib, HDAC inhibitors, mTOR inhibitors, ruxolitinib, and venetoclax, a detailed review of data available until August 2018 has been conducted, and specific recommendations for prophylaxis, diagnostic and differential diagnostic procedures as well as for clinical management have been developed.
Collapse
Affiliation(s)
- Georg Maschmeyer
- Department of Hematology, Oncology and Palliative Care, Klinikum Ernst von Bergmann, Charlottenstrasse 72, 14467, Potsdam, Germany.
| | - Julien De Greef
- Department of Internal Medicine and Infectious Diseases, Saint-Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Hematology, Henri Mondor Teaching Hospital, Créteil, France
| | - Sibylle C Mellinghoff
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Annamaria Nosari
- Department of Hematology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Anne Bergeron
- Department of Pneumology, Université Paris Diderot, APHP Saint-Louis Hospital, Paris, France
| | - Tomas Franquet
- Department of Radiology, Hospital de Sant Pau, Barcelona, Spain
| | | | - Johan A Maertens
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Dostert C, Grusdat M, Letellier E, Brenner D. The TNF Family of Ligands and Receptors: Communication Modules in the Immune System and Beyond. Physiol Rev 2019; 99:115-160. [DOI: 10.1152/physrev.00045.2017] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The tumor necrosis factor (TNF) and TNF receptor (TNFR) superfamilies (TNFSF/TNFRSF) include 19 ligands and 29 receptors that play important roles in the modulation of cellular functions. The communication pathways mediated by TNFSF/TNFRSF are essential for numerous developmental, homeostatic, and stimulus-responsive processes in vivo. TNFSF/TNFRSF members regulate cellular differentiation, survival, and programmed death, but their most critical functions pertain to the immune system. Both innate and adaptive immune cells are controlled by TNFSF/TNFRSF members in a manner that is crucial for the coordination of various mechanisms driving either co-stimulation or co-inhibition of the immune response. Dysregulation of these same signaling pathways has been implicated in inflammatory and autoimmune diseases, highlighting the importance of their tight regulation. Investigation of the control of TNFSF/TNFRSF activities has led to the development of therapeutics with the potential to reduce chronic inflammation or promote anti-tumor immunity. The study of TNFSF/TNFRSF proteins has exploded over the last 30 yr, but there remains a need to better understand the fundamental mechanisms underlying the molecular pathways they mediate to design more effective anti-inflammatory and anti-cancer therapies.
Collapse
Affiliation(s)
- Catherine Dostert
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Melanie Grusdat
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Elisabeth Letellier
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Dirk Brenner
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
20
|
Simo OC, Warren SJ, Mark L, Hoffmann K, Alomari AK. CD
8‐positive lymphomatoid papulosis (type D): Some lesions may lack
CD
30 expression and overlap histologically with mycosis fungoides. Int J Dermatol 2018; 58:800-805. [DOI: 10.1111/ijd.14309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/30/2018] [Accepted: 10/30/2018] [Indexed: 12/01/2022]
Affiliation(s)
- Olivia C. Simo
- School of Medicine Indiana University Indianapolis IN USA
| | - Simon J. Warren
- School of Medicine Indiana University Indianapolis IN USA
- Department of Pathology Indiana University Indianapolis IN USA
- Department of Dermatology Indiana University Indianapolis IN USA
| | - Lawrence Mark
- School of Medicine Indiana University Indianapolis IN USA
- Department of Dermatology Indiana University Indianapolis IN USA
| | - Kristin Hoffmann
- School of Medicine Indiana University Indianapolis IN USA
- Department of Dermatology Indiana University Indianapolis IN USA
| | - Ahmed K. Alomari
- School of Medicine Indiana University Indianapolis IN USA
- Department of Pathology Indiana University Indianapolis IN USA
- Department of Dermatology Indiana University Indianapolis IN USA
| |
Collapse
|
21
|
Soluble CD30, Acute Rejection, and Graft Survival: Pre- and 6-Month Post-Transplant Determinations-When Is the Best Time to Measure? Transplant Proc 2018; 50:728-736. [PMID: 29661425 DOI: 10.1016/j.transproceed.2018.02.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Pretransplantation soluble CD30 (sCD30) has been shown to be a good predictor of acute rejection (AR) and graft loss. This study aimed to evaluate the effectiveness of sCD30 measured pretransplant and up to 6 months after transplantation as a predictor of AR, graft loss, and survival at 5 years post-transplantation. Subjects were patients receiving living donor renal transplants at Bonsucesso Federal Hospital (Rio de Janeiro) in 2006 and between August 2010 and May 2011. METHODS sCD30 was analyzed in samples collected pretransplantation and 7, 14, and 21, 28 days and 3, 4, 5, and 6 months post-transplantation from 73 kidney recipients. RESULTS Patients in the AR group did not present a positive correlation with the sCD30 levels pretransplant (P = .54); in the post-transplant period, the 7- to 14-day samples showed patients with AR had higher levels of this biomarker (P = .036). The graft survival in 5 years of follow-up was not different between groups. CONCLUSIONS The best time to predict AR using sCD30 is the 7- to 14-day sample; however, identifying and following the decrease of this biomarker from pre- to post-transplant seems to be better than just 1 measurement. The sCD30 post-transplant is another tool that may be used in monitoring patients after renal transplantation.
Collapse
|
22
|
Nash AJ, Mandaviya PR, Dib MJ, Uitterlinden AG, van Meurs J, Heil SG, Andrew T, Ahmadi KR. Interaction between plasma homocysteine and the MTHFR c.677C > T polymorphism is associated with site-specific changes in DNA methylation in humans. FASEB J 2018; 33:833-843. [PMID: 30080444 DOI: 10.1096/fj.201800400r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
One-carbon metabolism provides a direct link among dietary folate/vitamin B12 exposure, the activity of the enzyme methylenetetrahydrofolate reductase (MTHFR), and epigenetic regulation of the genome via DNA methylation. Previously, it has been shown that the common c.677C > T polymorphism in MTHFR influences global DNA methylation status through a direct interaction with folate status and (indirectly) with total homocysteine (tHcy) levels. To build on that and other more recent observations that have further highlighted associations among MTHFR c.677C > T, tHcy, and aberrations in DNA methylation, we investigated whether the interaction between mildly elevated plasma tHcy and the c.677C > T polymorphism is associated with site-specific changes in DNA methylation in humans. We used data on plasma tHcy levels, c.677C > T polymorphism, and site-specific DNA methylation levels for a total of 915 white women and 335 men from the TwinsUK registry ( n = 610) and the Rotterdam study ( n = 670). We performed methylome-wide association analyses in each cohort to model the interaction between levels of tHcy and c.677C > T genotypes on DNA methylation β values. Our meta-analysis identified 13 probes significantly associated with rs1801133 × tHcy levels [false-discovery rate (FDR) < 0.05]. The most significant associations were with a cluster of probes at the AGTRAP-MTHFR-NPPA/B gene locus on chromosome 1 (FDR = 1.3E-04), with additional probes on chromosomes 2, 3, 4, 7, 12, 16, and 19. Our top 2 hits on chromosome 1 were functionally associated with variability in expression of the TNF receptor superfamily member 8 ( TNFRSF8) gene/locus on that chromosome. This is the first study, to our knowledge, to provide a direct link between perturbations in 1-carbon metabolism, through an interaction of tHcy and the activity of MTHFR enzyme on epigenetic regulation of the genome via DNA methylation.-Nash, A. J., Mandaviya, P. R., Dib, M.-J., Uitterlinden, A. G., van Meurs, J., Heil, S. G., Andrew, T., Ahmadi, K. R. Interaction between plasma homocysteine and the MTHFR c.677C>T polymorphism is associated with site-specific changes in DNA methylation in humans.
Collapse
Affiliation(s)
- Alexander J Nash
- Institute of Clinical Sciences and Medical Research Council (MRC) London Institute of Medical Sciences, Imperial College, London, United Kingdom
| | - Pooja R Mandaviya
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marie-Joe Dib
- Department of Genomics of Common Disease, Imperial College, London, United Kingdom; and
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Joyce van Meurs
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sandra G Heil
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Toby Andrew
- Department of Genomics of Common Disease, Imperial College, London, United Kingdom; and
| | - Kourosh R Ahmadi
- Department of Nutritional Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
23
|
Kang L, Jiang D, Ehlerding EB, Barnhart TE, Ni D, Engle JW, Wang R, Huang P, Xu X, Cai W. Noninvasive Trafficking of Brentuximab Vedotin and PET Imaging of CD30 in Lung Cancer Murine Models. Mol Pharm 2018. [PMID: 29537283 DOI: 10.1021/acs.molpharmaceut.7b01168] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
CD30 has been considered a unique diagnostic and therapeutic target for CD30-positive lymphomas and some lung diseases. Additionally, CD30 has shown high expression in clinical lung cancer samples. In this study, 89Zr-radiolabeled brentuximab vedotin (BV) was developed for in vivo tracking of BV and imaging CD30 expression in lung cancer models via conjugation with desferrioxamine (Df). CD30 expression in three lung cancer cell lines (H460, H358, and A549) was quantified by Western blot. Flow cytometry and saturation binding assays were used to evaluate the binding capabilities of the tracer in vitro. After longitudinal positron emission tomography (PET) imaging and quantitative analysis were performed, ex vivo biodistribution and histological studies were used to verify PET results. Finally, dosimetric extrapolation of murine data to humans was performed. At the cellular level, CD30 was found to be expressed on H460 and A549 cells with the highest and lowest levels of expression, respectively. Both Df-BV and 89Zr-Df-BV displayed high binding affinity to H460 cells. PET images and their quantification verified that BV accumulated in H460 tumor models (9.93 ± 2.70% ID/g at 24 h after injection; n = 4) at the highest level, followed by H358 and A549 tumors (8.05 ± 2.43 and 5.00 ± 1.56% ID/g; n = 4). The nonspecific 89Zr-labeled IgG showed a low tumor uptake of 5.2 ± 1.0% ID/g for H460 models. Ex vivo biodistribution and fluorescence immunohistochemistry also corroborated these findings. Dosimetric results displayed safe dose estimations. Therefore, 89Zr-Df-BV provides a potential agent for evaluating CD30 expression noninvasively in lung cancer, and also for imaging of brentuximab vedotin for better understanding of its pharmacokinetics.
Collapse
Affiliation(s)
- Lei Kang
- Department of Nuclear Medicine , Peking University First Hospital , Beijing 100034 , China.,Department of Radiology , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Dawei Jiang
- Department of Radiology , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States.,Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center , Shenzhen University , Shenzhen 518060 , China
| | - Emily B Ehlerding
- Department of Medical Physics , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Todd E Barnhart
- Department of Medical Physics , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Dalong Ni
- Department of Radiology , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Jonathan W Engle
- Department of Medical Physics , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Rongfu Wang
- Department of Nuclear Medicine , Peking University First Hospital , Beijing 100034 , China
| | - Peng Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center , Shenzhen University , Shenzhen 518060 , China
| | - Xiaojie Xu
- Department of Medical Molecular Biology , Beijing Institute of Biotechnology , Beijing 100850 , China
| | - Weibo Cai
- Department of Radiology , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States.,Department of Medical Physics , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States.,University of Wisconsin Carbone Cancer Center , Madison , Wisconsin 53705 , United States
| |
Collapse
|
24
|
Wang R, Li L, Zhang S, Li Y, Wang X, Miao Q, Zhen Y. A novel enediyne-integrated antibody-drug conjugate shows promising antitumor efficacy against CD30 + lymphomas. Mol Oncol 2018; 12:339-355. [PMID: 29316337 PMCID: PMC5830626 DOI: 10.1002/1878-0261.12166] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/25/2017] [Accepted: 12/26/2017] [Indexed: 01/26/2023] Open
Abstract
CD30 is a 120-kDa type I transmembrane glycoprotein belonging to the tumor necrosis factor receptor superfamily. Overexpression of CD30 has been reported in Hodgkin's lymphoma (HL) and anaplastic large-cell lymphoma (ALCL). CD30-targeted treatment with antibody-drug conjugates (ADCs) can lead to promising clinical benefit. Lidamycin (LDM), consisting of an apoprotein LDP and an active enediyne chromophore AE, is a member of the enediyne antibiotic family and one of the most potent antitumor agents. AE and LDP can be dissociated and reconstituted under certain conditions in vitro. LDM is an ideal payload for the preparation of ADCs. In this study, we show the generation, production, and antitumor activity of anti-CD30-LDM, a novel ADC which consists of the intact anti-CD30 antibody and LDM. First, the anti-CD30-LDP fusion protein was constructed and expressed in CHO/dhFr- cells. Anti-CD30-LDP showed specific and high-affinity binding to CD30 and could be internalized into target cells. It also exhibited excellent tumor-targeting capability in vivo. Next, anti-CD30-LDM was prepared by assembling the enediyne molecule AE to the fusion protein anti-CD30-LDP. Anti-CD30-LDM was highly cytotoxic to HL and ALCL cell lines, with IC50 values of 5-50 pm. It can also induce cell apoptosis and G2/M cell cycle arrest. In the Karpas299 xenograft model, the tumor growth was inhibited by 87.76% in mice treated with anti-CD30-LDM and with no discernible adverse effects. Taken together, anti-CD30-LDM shows attractive tumor-targeting capability and antitumor efficacy both in vitro and in vivo and could be a promising candidate for the treatment of CD30+ lymphomas.
Collapse
Affiliation(s)
- Rong Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Liang Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shenghua Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yi Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaofei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Qingfang Miao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yongsu Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
Zhang LL, Wei JY, Wang L, Huang SL, Chen JL. Human T-cell lymphotropic virus type 1 and its oncogenesis. Acta Pharmacol Sin 2017; 38:1093-1103. [PMID: 28392570 PMCID: PMC5547553 DOI: 10.1038/aps.2017.17] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 02/27/2017] [Indexed: 02/08/2023]
Abstract
Human T-cell lymphotropic virus type 1 (HTLV-1) is the etiologic agent of adult T-cell leukemia/lymphoma (ATL), a rapidly progressing clonal malignancy of CD4+ T lymphocytes. Exploring the host-HTLV-1 interactions and the molecular mechanisms underlying HTLV-1-mediated tumorigenesis is critical for developing efficient therapies against the viral infection and associated leukemia/lymphoma. It has been demonstrated to date that several HTLV-1 proteins play key roles in the cellular transformation and immortalization of infected T lymphocytes. Of note, the HTLV-1 oncoprotein Tax inhibits the innate IFN response through interaction with MAVS, STING and RIP1, causing the suppression of TBK1-mediated phosphorylation of IRF3/IRF7. The HTLV-1 protein HBZ disrupts genomic integrity and inhibits apoptosis and autophagy of the target cells. Furthermore, it is revealed that HBZ enhances the proliferation of ATL cells and facilitates evasion of the infected cells from immunosurveillance. These studies provide insights into the molecular mechanisms by which HTLV-1 mediates the formation of cancer as well as useful strategies for the development of new therapeutic interventions against ATL. In this article, we review the recent advances in the understanding of the pathogenesis, the underlying mechanisms, clinical diagnosis and treatment of the disease caused by HTLV-1 infection. In addition, we discuss the future direction for targeting HTLV-1-associated cancers and strategies against HTLV-1.
Collapse
Affiliation(s)
- Lan-lan Zhang
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jing-yun Wei
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Long Wang
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Shi-le Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Ji-long Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
26
|
Abstract
Primary cutaneous CD30+ lymphoproliferative disorders encompass lymphomatoid papulosis (LyP), primary cutaneous anaplastic large cell lymphoma (pcALCL), and indeterminate cases. LyP is a benign disorder characterized by recurrent crops of red or violaceous papulonodules. Patients with LyP are at an increased risk of a secondary malignancy. pcALCL is characterized by a solitary red to violaceous nodule or tumor larger than 20 mm. LyP is benign, is limited to the skin, and self-resolves, with a 5-year survival rate of 100%; pcALCL is limited to the skin and responsive to directed therapies, with a 5-year survival rate of over 95%. Aggressive chemotherapeutic regimens should be avoided.
Collapse
MESH Headings
- Disease-Free Survival
- Humans
- Lymphoma, Large-Cell, Anaplastic/diagnosis
- Lymphoma, Large-Cell, Anaplastic/drug therapy
- Lymphoma, Large-Cell, Anaplastic/metabolism
- Lymphoma, Large-Cell, Anaplastic/mortality
- Lymphomatoid Papulosis/diagnosis
- Lymphomatoid Papulosis/drug therapy
- Lymphomatoid Papulosis/metabolism
- Lymphomatoid Papulosis/mortality
- Neoplasms, Second Primary/diagnosis
- Neoplasms, Second Primary/drug therapy
- Neoplasms, Second Primary/metabolism
- Neoplasms, Second Primary/mortality
- Risk Factors
- Skin Neoplasms/diagnosis
- Skin Neoplasms/drug therapy
- Skin Neoplasms/metabolism
- Skin Neoplasms/microbiology
- Survival Rate
Collapse
Affiliation(s)
- Maxwell B Sauder
- Department of Dermatology, The Center for Cutaneous Oncology, Dana Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02115, USA
| | - John T O'Malley
- Department of Dermatology, The Center for Cutaneous Oncology, Dana Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02115, USA
| | - Nicole R LeBoeuf
- Department of Dermatology, The Center for Cutaneous Oncology, Dana Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Abstract
INTRODUCTION CD30 is a cell surface receptor expressed in classical Hodgkin lymphoma (HL), anaplastic large cell lymphoma (ALCL), and many other lymphomas to a variable degree. It has been identified as an important therapeutic target in lymphoma. Areas covered: CD30 testing is essential in diagnosis of classical HL and ALCL, and expression can also be seen in other lymphoma subtypes. Development of Brentuximab vedotin (BV), an antibody-drug conjugate directed to CD30, has been an important advance in lymphoma treatment. It is approved in treatment of relapsed HL and ALCL, as well as post-transplant maintenance for HL, and has been shown to be effective in other CD30-expressing lymphomas. This review describes the role of CD30 and the use of CD30-targeted agents in HL, ALCL, and other lymphomas, including review of relevant trials of BV. Expert commentary: Recognition of CD30 expression in lymphoma has led to the development of important therapeutic options. Multiple trials are ongoing combining BV with other agents, such as chemotherapy or immunotherapy, to develop more effective regimens. In addition, treatments targeting CD30 in different ways are being developed, such as bispecific antibodies and chimeric antigen receptor (CAR) T-cells.
Collapse
Affiliation(s)
- John Matthew R Pierce
- a Hematology & Oncology , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Amitkumar Mehta
- a Hematology & Oncology , University of Alabama at Birmingham , Birmingham , AL , USA.,b Department Of Medicine , University of Alabama at Birmingham , Birmingham , AL , USA
| |
Collapse
|
28
|
Vallacchi V, Camisaschi C, Dugo M, Vergani E, Deho P, Gualeni A, Huber V, Gloghini A, Maurichi A, Santinami M, Sensi M, Castelli C, Rivoltini L, Rodolfo M. microRNA Expression in Sentinel Nodes from Progressing Melanoma Patients Identifies Networks Associated with Dysfunctional Immune Response. Genes (Basel) 2016; 7:genes7120124. [PMID: 27983661 PMCID: PMC5192500 DOI: 10.3390/genes7120124] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 11/24/2016] [Accepted: 12/05/2016] [Indexed: 12/21/2022] Open
Abstract
Sentinel node biopsy (SNB) is a main staging biomarker in melanoma and is the first lymph node to drain the tumor, thus representing the immunological site where anti-tumor immune dysfunction is established and where potential prognostic immune markers can be identified. Here we analyzed microRNA (miR) profiles in archival tumor-positive SNBs derived from melanoma patients with different outcomes and performed an integrated analysis of transcriptional data to identify deregulated immune signaling networks. Twenty-six miRs were differentially expressed in melanoma-positive SNB samples between patients with disease progression and non-progressing patients, the majority being previously reported in the regulation of immune responses. A significant variation in miR expression levels was confirmed in an independent set of SNB samples. Integrated information from genome-wide transcriptional profiles and in vitro assessment in immune cells led to the identification of miRs associated with the regulation of the TNF receptor superfamily member 8 (TNFRSF8) gene encoding the CD30 receptor, a marker increased in lymphocytes of melanoma patients with progressive disease. These findings indicate that miRs are involved in the regulation of pathways leading to immune dysfunction in the sentinel node and may provide valuable markers for developing prognostic molecular signatures for the identification of stage III melanoma patients at risk of recurrence.
Collapse
Affiliation(s)
- Viviana Vallacchi
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Chiara Camisaschi
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Matteo Dugo
- Functional Genomics and Bioinformatics, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Elisabetta Vergani
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Paola Deho
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Ambra Gualeni
- Molecular Pathology Unit, Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Veronica Huber
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Annunziata Gloghini
- Molecular Pathology Unit, Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Andrea Maurichi
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Mario Santinami
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Marialuisa Sensi
- Functional Genomics and Bioinformatics, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Chiara Castelli
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Licia Rivoltini
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| | - Monica Rodolfo
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy.
| |
Collapse
|
29
|
Berger GK, McBride A, Lawson S, Royball K, Yun S, Gee K, Bin Riaz I, Saleh AA, Puvvada S, Anwer F. Brentuximab vedotin for treatment of non-Hodgkin lymphomas: A systematic review. Crit Rev Oncol Hematol 2016; 109:42-50. [PMID: 28010897 DOI: 10.1016/j.critrevonc.2016.11.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/12/2016] [Accepted: 11/17/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Brentuximab vedotin (BV) is an antibody-drug conjucate (ADC) comprising a CD30-directed antibody, conjugated to the microtubule-disrupting agent MMAE via a protease cleavable linker. BV is FDA approved for use in relapsed classical Hodgkin lymphoma (HL) and relapsed systemic anaplastic large cell lymphoma (sALCL). There are multiple publications for its utility in other malignancies such as diffuse large B-cell lymphoma (DLBCL), mycosis fungoides (MF), Sézary syndrome (SS), T-cell lymphomas (TCL), primary mediastinal lymphoma (PMBL), and post-transplant lymphoproliferative disorders (PTLD). We believe that BV could potentially provide a strong additional treatment option for patients suffering from NHL. OBJECTIVE Perform a systematic review on the use of BV in non-Hodgkin lymphoma (NHL) and other CD30+ malignancies in humans. DATA SOURCES We searched various databases including PubMed (1946-2015), EMBASE (1947-2015), and Cochrane Central Register of Controlled Trials (1898-2015). ELIGIBILITY CRITERIA Inclusion criteria specified all studies and case reports of NHLs in which BV therapy was administered. INCLUDED STUDIES A total of 28 articles met these criteria and are summarized in this manuscript. CONCLUSION Our findings indicate that BV induces a variety of responses, largely positive in nature and variable between NHL subtypes. With additional, properly powered prospective studies, BV may prove to be a strong candidate in the treatment of various CD30+ malignancies.
Collapse
Affiliation(s)
- Garrett K Berger
- College of Pharmacy, University of Arizona, Tucson, AZ, 85721, United States
| | - Ali McBride
- College of Pharmacy, University of Arizona, Tucson, AZ, 85721, United States
| | - Stephanie Lawson
- College of Pharmacy, University of Arizona, Tucson, AZ, 85721, United States
| | - Kelsey Royball
- College of Pharmacy, University of Arizona, Tucson, AZ, 85721, United States
| | - Seongseok Yun
- Departments of Medicine, University of Arizona, Tucson, AZ, 85721, United States
| | - Kevin Gee
- College of Medicine, University of Arizona, Tucson, AZ, 85721, United States
| | - Irbaz Bin Riaz
- Departments of Medicine, University of Arizona, Tucson, AZ, 85721, United States
| | - Ahlam A Saleh
- University of Arizona Health Sciences Library, University of Arizona, Tucson, AZ 85721, United States
| | - Soham Puvvada
- Departments of Medicine, University of Arizona, Tucson, AZ, 85721, United States
| | - Faiz Anwer
- Hematology, Oncology, Blood & Marrow Transplantation, Department of Medicine, University of Arizona, Tucson, AZ, 85721, United States.
| |
Collapse
|
30
|
Abstract
Through years of evolutionary selection pressures, organisms have developed potent toxins that coincidentally have marked antineoplastic activity. These natural products have been vital for the development of multiagent treatment regimens currently employed in cancer chemotherapy, and are used in the treatment of a variety of malignancies. Therefore, this review catalogs recent advances in natural product-based drug discovery via the examination of mechanisms of action and available clinical data to highlight the utility of these novel compounds in the burgeoning age of precision medicine. The review also highlights the recent development of antibody-drug conjugates and other immunotoxins, which are capable of delivering highly cytotoxic agents previously deemed too toxic to elicit therapeutic benefit preferentially to neoplastic cells. Finally, the review examines natural products not currently used in the clinic that have novel mechanisms of action, and may serve to supplement current chemotherapeutic protocols.
Collapse
|
31
|
Bhatt G, Maddocks K, Christian B. CD30 and CD30-Targeted Therapies in Hodgkin Lymphoma and Other B cell Lymphomas. Curr Hematol Malig Rep 2016; 11:480-491. [DOI: 10.1007/s11899-016-0345-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
32
|
Hombach AA, Abken H. Shared target antigens on cancer cells and tissue stem cells: go or no-go for CAR T cells? Expert Rev Clin Immunol 2016; 13:151-155. [PMID: 27546707 DOI: 10.1080/1744666x.2016.1221763] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Adoptive therapy with chimeric antigen receptor (CAR) T cells redirected towards CD19 produces remissions of B cell malignancies, however, it also eradicates healthy B cells sharing the target antigen. Such 'on-target off-tumor' toxicity raises serious safety concerns when the target antigen is also expressed by tissue stem cells, with the risk of lasting tissue destruction. Areas covered: We discuss CAR T cell targeting of activation antigens versus lineage associated antigens on the basis of recent experimental and animal data and the literature in the field. Expert commentary: Targeting an activation associated antigen which is transiently expressed by stem cells seems to be safe, like CAR T cells targeting CD30 spare CD30+ hematopoietic stem and progenitor cells while eliminating CD30+ lymphoma cells, whereas targeting lineage associated antigens which increase in expression during cell maturation, like folate receptor-β and CD123, is of risk to destruct tissue stem cells.
Collapse
Affiliation(s)
- Andreas A Hombach
- a Center for Molecular Medicine Cologne , University of Cologne , Cologne , Germany.,b Department I of Internal Medicine , University Hospital Cologne , Cologne , Germany
| | - Hinrich Abken
- a Center for Molecular Medicine Cologne , University of Cologne , Cologne , Germany.,b Department I of Internal Medicine , University Hospital Cologne , Cologne , Germany
| |
Collapse
|
33
|
Superior Therapeutic Index in Lymphoma Therapy: CD30(+) CD34(+) Hematopoietic Stem Cells Resist a Chimeric Antigen Receptor T-cell Attack. Mol Ther 2016; 24:1423-34. [PMID: 27112062 DOI: 10.1038/mt.2016.82] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 03/16/2016] [Indexed: 12/17/2022] Open
Abstract
Recent clinical trials with chimeric antigen receptor (CAR) redirected T cells targeting CD19 revealed particular efficacy in the treatment of leukemia/lymphoma, however, were accompanied by a lasting depletion of healthy B cells. We here explored CD30 as an alternative target, which is validated in lymphoma therapy and expressed by a broad variety of Hodgkin's and non-Hodgkin's lymphomas. As a safty concern, however, CD30 is also expressed by lymphocytes and hematopoietic stem and progenitor cells (HSPCs) during activation. We revealed that HRS3scFv-derived CAR T cells are superior since they were not blocked by soluble CD30 and did not attack CD30(+) HSPCs while eliminating CD30(+) lymphoma cells. Consequently, normal hemato- and lymphopoiesis was not affected in the long-term in the humanized mouse; the number of blood B and T cells remained unchanged. We provide evidence that the CD30(+) HSPCs are protected against a CAR T-cell attack by substantially lower CD30 levels than lymphoma cells and higher levels of the granzyme B inactivating SP6/PI9 serine protease, which furthermore increased upon activation. Taken together, adoptive cell therapy with anti-CD30 CAR T cells displays a superior therapeutic index in the treatment of CD30(+) malignancies leaving healthy activated lymphocytes and HSPCs unaffected.
Collapse
|
34
|
Raja SM, Desale SS, Mohapatra B, Luan H, Soni K, Zhang J, Storck MA, Feng D, Bielecki TA, Band V, Cohen SM, Bronich TK, Band H. Marked enhancement of lysosomal targeting and efficacy of ErbB2-targeted drug delivery by HSP90 inhibition. Oncotarget 2016; 7:10522-35. [PMID: 26859680 PMCID: PMC4891137 DOI: 10.18632/oncotarget.7231] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 01/26/2016] [Indexed: 12/13/2022] Open
Abstract
Targeted delivery of anticancer drugs to tumor cells using monoclonal antibodies against oncogenic cell surface receptors is an emerging therapeutic strategy. These strategies include drugs directly conjugated to monoclonal antibodies through chemical linkers (Antibody-Drug Conjugates, ADCs) or those encapsulated within nanoparticles that in turn are conjugated to targeting antibodies (Antibody-Nanoparticle Conjugates, ANPs). The recent FDA approval of the ADC Trastuzumab-TDM1 (Kadcyla; Genentech; San Francisco) for the treatment of ErbB2-overexpressing metastatic breast cancer patients has validated the strong potential of these strategies. Even though the activity of ANPs and ADCs is dependent on lysosomal traffic, the roles of the endocytic route traversed by the targeted receptor and of cancer cell-specific alterations in receptor dynamics on the efficiency of drug delivery have not been considered in these new targeted therapies. For example, constitutive association with the molecular chaperone HSP90 is thought to either retard ErbB2 endocytosis or to promote its recycling, traits undesirable for targeted therapy with ANPs and ADCs. HSP90 inhibitors are known to promote ErbB2 ubiquitination, targeting to lysosome and degradation. We therefore hypothesized that ErbB2-targeted drug delivery using Trastuzumab-conjugated nanoparticles could be significantly improved by HSP90 inhibitor-promoted lysosomal traffic of ErbB2. Studies reported here validate this hypothesis and demonstrate, both in vitro and in vivo, that HSP90 inhibition facilitates the intracellular delivery of Trastuzumab-conjugated ANPs carrying a model chemotherapeutic agent, Doxorubicin, specifically into ErbB2-overexpressing breast cancer cells, resulting in improved antitumor activity. These novel findings highlight the need to consider oncogene-specific alterations in receptor traffic in the design of targeted drug delivery strategies. We suggest that combination of agents that enhance receptor endocytosis and lysosomal routing can provide a novel strategy to significantly improve therapy with ANPs and ADCs.
Collapse
Affiliation(s)
- Srikumar M. Raja
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Swapnil S. Desale
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, Omaha, Nebraska, USA
| | - Bhopal Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA
| | - Haitao Luan
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA
| | - Kruti Soni
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, Omaha, Nebraska, USA
| | - Jinjin Zhang
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, Omaha, Nebraska, USA
| | - Matthew A. Storck
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA
| | - Dan Feng
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA
| | - Timothy A. Bielecki
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA
| | - Vimla Band
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Samuel M. Cohen
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tatiana K. Bronich
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, Omaha, Nebraska, USA
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Departments of Biochemistry and Molecular Biology, Pathology and Microbiology and Pharmacology and Neuroscience, College of Medicine, Omaha, Nebraska, USA
| |
Collapse
|
35
|
Lisenko K, Schönland S, Hegenbart U, Wallenwein K, Braun U, Mai EK, Hillengass J, Goldschmidt H, Jauch A, Ho AD, Raab M, Hundemer M. Potential therapeutic targets in plasma cell disorders: A flow cytometry study. CYTOMETRY PART B-CLINICAL CYTOMETRY 2016; 92:145-152. [DOI: 10.1002/cyto.b.21351] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/03/2015] [Accepted: 12/08/2015] [Indexed: 02/05/2023]
Affiliation(s)
- Katharina Lisenko
- Department of Internal Medicine V; University of Heidelberg; Heidelberg Germany
| | - Stefan Schönland
- Department of Internal Medicine V; University of Heidelberg; Heidelberg Germany
| | - Ute Hegenbart
- Department of Internal Medicine V; University of Heidelberg; Heidelberg Germany
| | - Katrin Wallenwein
- Department of Internal Medicine V; University of Heidelberg; Heidelberg Germany
| | - Ute Braun
- Department of Internal Medicine V; University of Heidelberg; Heidelberg Germany
| | - Elias K. Mai
- Department of Internal Medicine V; University of Heidelberg; Heidelberg Germany
| | - Jens Hillengass
- Department of Internal Medicine V; University of Heidelberg; Heidelberg Germany
| | - Hartmut Goldschmidt
- Department of Internal Medicine V; University of Heidelberg; Heidelberg Germany
- National Center for Tumor Diseases, University of Heidelberg; Heidelberg Germany
| | - Anna Jauch
- Institute of Human Genetics, University of Heidelberg; Heidelberg Germany
| | - Anthony D. Ho
- Department of Internal Medicine V; University of Heidelberg; Heidelberg Germany
| | - Marc Raab
- Department of Internal Medicine V; University of Heidelberg; Heidelberg Germany
| | - Michael Hundemer
- Department of Internal Medicine V; University of Heidelberg; Heidelberg Germany
| |
Collapse
|
36
|
Takemoto S, Iwanaga M, Sagara Y, Watanabe T. Plasma Soluble CD30 as a Possible Marker of Adult T-cell Leukemia in HTLV-1 Carriers: a Nested Case-Control Study. Asian Pac J Cancer Prev 2016; 16:8253-8. [DOI: 10.7314/apjcp.2015.16.18.8253] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
37
|
Rylova SN, Del Pozzo L, Klingeberg C, Tönnesmann R, Illert AL, Meyer PT, Maecke HR, Holland JP. Immuno-PET Imaging of CD30-Positive Lymphoma Using 89Zr-Desferrioxamine-Labeled CD30-Specific AC-10 Antibody. J Nucl Med 2015; 57:96-102. [PMID: 26514172 DOI: 10.2967/jnumed.115.162735] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/13/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The CD30-specific antibody-drug conjugate, brentuximab vedotin, is approved for the treatment of relapsed, refractory Hodgkin lymphomas and systemic anaplastic large T-cell lymphomas. Multiple ongoing clinical trials are investigating brentuximab vedotin efficacy in other CD30-positive hematologic malignancies. Because CD30 expression varies among different types of lymphoma and can also change during the course of treatment, companion diagnostic imaging of CD30 could be a valuable tool in optimizing patient-specific brentuximab vedotin treatment regimens. METHODS The mouse antihuman CD30 antibody AC-10 was radiolabeled with the positron-emitting radionuclide (89)Zr. The stability and specificity of (89)Zr-desferrioxamine (DFO)-labeled CD30-specific AC-10 antibody ((89)Zr-DFO-AC-10) was evaluated in vitro. The pharmacokinetics of (89)Zr-DFO-AC-10 was studied in BALB/c nude mice bearing subcutaneous human Karpas 299 tumors (CD30-positive model) or A-431 tumors (CD30-negative model) using PET/CT imaging, biodistribution studies, and autoradiography. RESULTS AC-10 was conjugated with a DFO B chelator and radiolabeled with (89)Zr to give formulated (89)Zr-DFO-AC-10 with a radiochemical yield of 80%, radiochemical purity greater than 99%, and specific activity of 111-148 MBq/mg. (89)Zr-DFO-AC-10 was stable in mouse and human sera and preserved the immunoreactivity toward CD30. Biodistribution data showed the highest tissue accumulation of (89)Zr-DFO-AC-10 in CD30-positive tumors, with 37.9% ± 8.2% injected activity per gram of tissue at 72 h after injection, whereas uptake in CD30-negative tumors was 11.0% ± 0.4%. The specificity of (89)Zr-DFO-AC-10 binding to CD30 in vivo was confirmed by blocking studies. Time-activity curves showed that between 24 and 144 h after injection, tumor-to-muscle ratios increased from 18.9 to 51.8 in the CD30-positive model and from 4.8 to 8.7 in the CD30-negative model. Tumor-to-blood ratios also increased, from 3.2 to 13.6 and from 1 to 2 in the CD30-positive and -negative models, respectively. CONCLUSION Our results demonstrate that for measuring CD30 expression, (89)Zr-DFO-AC-10 is a sensitive PET agent with high tumor-to-normal-tissue contrast. (89)Zr-DFO-AC-10 is a promising CD30-imaging radiotracer for clinical translation in patients with various lymphomas and other diseases.
Collapse
Affiliation(s)
- Svetlana N Rylova
- German Cancer Consortium, Heidelberg, Germany Department of Nuclear Medicine, University Hospital Freiburg, Freiburg, Germany German Cancer Research Center, Heidelberg, Germany; and
| | - Luigi Del Pozzo
- German Cancer Consortium, Heidelberg, Germany Department of Nuclear Medicine, University Hospital Freiburg, Freiburg, Germany German Cancer Research Center, Heidelberg, Germany; and
| | - Cathrin Klingeberg
- German Cancer Consortium, Heidelberg, Germany German Cancer Research Center, Heidelberg, Germany; and Department of Internal Medicine I, University Hospital Freiburg, Freiburg, Germany
| | - Roswitha Tönnesmann
- Department of Nuclear Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Anna L Illert
- German Cancer Consortium, Heidelberg, Germany German Cancer Research Center, Heidelberg, Germany; and Department of Internal Medicine I, University Hospital Freiburg, Freiburg, Germany
| | - Philipp T Meyer
- German Cancer Consortium, Heidelberg, Germany Department of Nuclear Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Helmut R Maecke
- Department of Nuclear Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Jason P Holland
- German Cancer Consortium, Heidelberg, Germany Department of Nuclear Medicine, University Hospital Freiburg, Freiburg, Germany German Cancer Research Center, Heidelberg, Germany; and
| |
Collapse
|
38
|
Brentuximab vedotin demonstrates objective responses in a phase 2 study of relapsed/refractory DLBCL with variable CD30 expression. Blood 2015; 125:1394-402. [DOI: 10.1182/blood-2014-09-598763] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Key PointsBrentuximab vedotin was active in DLBCL across a range of CD30 expression levels, and objective responses occurred in 44% of patients.
Collapse
|
39
|
Rodolfo M, Castelli C, Rivoltini L. Immune response markers in sentinel nodes may predict melanoma progression. Oncoimmunology 2014; 3:e28498. [PMID: 25050216 PMCID: PMC4063153 DOI: 10.4161/onci.28498] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 03/10/2014] [Indexed: 11/19/2022] Open
Abstract
We recently reported that variable expression of immune-response genes distinguishes tumor positive sentinel nodes in melanoma patients with malignant progression from those with non-progressing disease. Our results depict sentinel nodes as sites in which immune functions are associated with metastatic disease and identify CD30 as a host immune-related cancer prognostic marker and potential therapeutic target.
Collapse
Affiliation(s)
- Monica Rodolfo
- Unit of Immunotherapy; Fondazione IRCCS Istituto Nazionale Tumori; Milan, Italy
| | - Chiara Castelli
- Unit of Immunotherapy; Fondazione IRCCS Istituto Nazionale Tumori; Milan, Italy
| | - Licia Rivoltini
- Unit of Immunotherapy; Fondazione IRCCS Istituto Nazionale Tumori; Milan, Italy
| |
Collapse
|