1
|
Abudureheman T, Zhou H, Yang LT, Huang XS, Jing JJ, Duan CW, Chen KM. Construction of Switch Modules for CAR-T Cell Treatment Using a Site-Specific Conjugation System. Bioconjug Chem 2024; 35:604-615. [PMID: 38661725 DOI: 10.1021/acs.bioconjchem.4c00050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Chimeric antigen receptor T-cell (CAR-T cell) therapy has become a promising treatment option for B-cell hematological tumors. However, few optional target antigens and disease relapse due to loss of target antigens limit the broad clinical applicability of CAR-T cells. Here, we conjugated an antibody (Ab) fusion protein, consisting of an Ab domain and a SpyCatcher domain, with the FITC-SpyTag (FITC-ST) peptide to form a bispecific safety switch module using a site-specific conjugation system. We applied the safety switch module to target CD19, PDL1, or Her2-expressing tumor cells by constructing FMC63 (anti-CD19), antiPDL1, or ZHER (anti-Her2)-FITC-ST, respectively. Those switch modules significantly improved the cytotoxic effects of anti-FITC CAR-T cells on tumor cells. Additionally, we obtained the purified CD8+ T cells by optimizing a shorter version of the CD8-binding aptamer to generate anti-FITC CD8-CAR-T cells, which combined with the CD4-FITC-ST switch module (anti-CD4) to eliminate the CD4-positive tumor cells in vitro and in vivo. Overall, we established a novel safety switch module by site-specific conjugation to enhance the antitumor function of universal CAR-T cells, thereby expanding the application scope of CAR-T therapy and improving its safety and efficacy.
Collapse
Affiliation(s)
- Tuersunayi Abudureheman
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hang Zhou
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Li-Ting Yang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xiu-Song Huang
- Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Jun-Jie Jing
- Fujian Branch of Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, and Fujian Children's Hospital, Fuzhou 350001, China
| | - Cai-Wen Duan
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
- Fujian Branch of Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, and Fujian Children's Hospital, Fuzhou 350001, China
| | - Kai-Ming Chen
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
- Fujian Branch of Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, and Fujian Children's Hospital, Fuzhou 350001, China
| |
Collapse
|
2
|
Feng J, Xu H, Cinquina A, Wu Z, Zhang W, Sun L, Chen Q, Tian L, Song L, Pinz KG, Wada M, Jiang X, Hanes WM, Ma Y, Zhang H. Treatment of aggressive T-cell lymphoma/leukemia with anti-CD4 CAR T cells. Front Immunol 2022; 13:997482. [PMID: 36172388 PMCID: PMC9511023 DOI: 10.3389/fimmu.2022.997482] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
T-cell lymphomas are aggressive lymphomas that often resist current therapy options or present with relapsed disease, making the development of more effective treatment regimens clinically important. Previously, we have shown that CD4 CAR can effectively target T-cell malignancies in preclinical studies. As IL-15 has been shown to strengthen the anti-tumor response, we have modified CD4 CAR to secrete an IL-15/IL-15sushi complex. These CD4-IL15/IL15sushi CAR T cells and NK92 cells efficiently eliminated CD4+ leukemic cell lines in co-culture assays. Additionally, CD4-IL15/IL15sushi CAR out-performed CD4 CAR in in vivo models, demonstrating a benefit to IL-15/IL-15sushi inclusion. In a Phase I clinical trial, CD4-IL15/IL15sushi CAR T cells were tested for safety in three patients with different T-cell lymphomas. Infusion of CD4-IL15/IL15sushi CAR T cells was well-tolerated by the patients without significant adverse effects and led to the remission of their lymphomas. Additionally, infusion led to the depletion of CD4+ Treg cells and expansion of CD3+CD8+ T cells and NK cells. These results suggest that CD4-IL15/IL15sushi CAR T cells may be a safe and effective treatment for patients with relapsed or refractory T-cell lymphomas, where new treatment options are needed.
Collapse
Affiliation(s)
- Jia Feng
- Department of Hematology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Haichan Xu
- Department of Hematology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Andrew Cinquina
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY, United States
| | - Zehua Wu
- Department of Hematology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wenli Zhang
- Department of Hematology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lihua Sun
- Department of Hematology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qi Chen
- Department of Hematology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lei Tian
- Department of Hematology, Peking University Third Hospital, Beijing, China
| | - Le Song
- Department of Nuclear Medicine, Peking University Third Hospital, Beijing, China
| | - Kevin G. Pinz
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY, United States
| | - Masayuki Wada
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY, United States
| | - Xun Jiang
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY, United States
| | - William M. Hanes
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY, United States
| | - Yupo Ma
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY, United States
- *Correspondence: Hongyu Zhang, ; Yupo Ma,
| | - Hongyu Zhang
- Department of Hematology, Peking University Shenzhen Hospital, Shenzhen, China
- *Correspondence: Hongyu Zhang, ; Yupo Ma,
| |
Collapse
|
3
|
Ramos-Tomillero I, Perez-Chacon G, Somovilla-Crespo B, Sanchez-Madrid F, Domínguez JM, Cuevas C, Zapata JM, Rodríguez H, Albericio F. Bioconjugation through Mesitylene Thiol Alkylation. Bioconjug Chem 2018; 29:1199-1208. [DOI: 10.1021/acs.bioconjchem.7b00828] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Iván Ramos-Tomillero
- Institute for Research in Biomedicine, 08028-Barcelona, Spain
- Department of Organic Chemistry, University of Barcelona, 08028-Barcelona, Spain
| | - Gema Perez-Chacon
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, 28029-Madrid, Spain
| | - Beatriz Somovilla-Crespo
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital de la Princesa, 28006-Madrid, Spain
| | - Francisco Sanchez-Madrid
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital de la Princesa, 28006-Madrid, Spain
| | | | - Carmen Cuevas
- Research Department, PharmaMar S.A., Colmenar Viejo, 28770-Madrid, Spain
| | - Juan Manuel Zapata
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, 28029-Madrid, Spain
| | | | - Fernando Albericio
- Institute for Research in Biomedicine, 08028-Barcelona, Spain
- Department of Organic Chemistry, University of Barcelona, 08028-Barcelona, Spain
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, 08028-Barcelona, Spain
- School of Chemistry, University of KwaZulu-Natal, 4001-Durban, South Africa
| |
Collapse
|
4
|
|
5
|
Fujinami N, Yoshikawa T, Sawada Y, Shimomura M, Iwama T, Sugai S, Kitano S, Uemura Y, Nakatsura T. Enhancement of antitumor effect by peptide vaccine therapy in combination with anti-CD4 antibody: Study in a murine model. Biochem Biophys Rep 2016; 5:482-491. [PMID: 28955856 PMCID: PMC5600353 DOI: 10.1016/j.bbrep.2016.02.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/17/2016] [Accepted: 02/17/2016] [Indexed: 01/30/2023] Open
Abstract
Purpose The clinical efficacy of cancer peptide vaccine therapy is insufficient. To enhance the anti-tumor effect of peptide vaccine therapy, we combined this therapy with an anti-CD4 mAb (GK1.5), which is known to deplete CD4+ cells, including regulatory T cells (Tregs). Methods To determine the treatment schedule, the number of lymphocyte subsets in the peripheral blood of mice was traced by flow cytometry after administration of anti-CD4 mAb. The ovalbumin (OVA)257–264 peptide vaccine was injected intradermally and anti-CD4 mAb was administered intraperitoneally into C57BL/6 mice at different schedules. We evaluated the enhancement of OVA peptide-specific cytotoxic T lymphocyte (CTL) induction in the combination therapy using the ELISPOT assay, CD107a assay, and cytokine assay. We then examined the in vivo metastasis inhibitory effect by OVA peptide vaccine therapy in combination with anti-CD4 mAb against OVA-expressing thymoma (EG7) in a murine liver metastatic model. Results We showed that peptide-specific CTL induction was enhanced by the peptide vaccine in combination with anti-CD4 mAb and that the optimized treatment schedule had the strongest induction effect of peptide-specific CTLs using an IFN-γ ELISPOT assay. We also confirmed that the CD107a+ cells secreted perforin and granzyme B and the amount of IL-2 and TNF produced by these CTLs increased when the peptide vaccine was combined with anti-CD4 mAb. Furthermore, metastasis was inhibited by peptide vaccines in combination with anti-CD4 mAb compared to peptide vaccine alone in a murine liver metastatic model. Conclusion The use of anti-CD4 mAb in combination with the OVA peptide vaccine therapy increased the number of peptide-specific CTLs and showed a higher therapeutic effect against OVA-expressing tumors. The combination with anti-CD4 mAb may provide a new cancer vaccine strategy. Peptide-specific CTL induction and function were enhanced by depletion of CD4+ cells. Anti-tumor effect by the peptide vaccine was enhanced by the depletion of CD4+ cells. Metastasis was inhibited by vaccine with depletion of CD4+ cells in a murine model. Combination with the depletion of CD4+ cells could be a new cancer vaccine strategy.
Collapse
Key Words
- 7-AAD, 7-amino-actinomycin D
- Anti-CD4 antibody
- CTL, cytotoxic T lymphocyte
- Cancer
- DC, dendritic cell
- ELISPOT assay, enzyme-linked immunospot assay
- FITC, fluorescein isothiocyanate
- FOXP3, forkhead box P3
- GPC3, glypican-3
- HCC, hepatocellular carcinoma
- IFN-γ, interferon-γ
- IL-2, interleukine-2
- Immunotherapy
- MHC, major histocompatibility complex
- Murine liver metastatic model
- OVA, ovalbumin
- PD-1, programmed death-1
- PE, phycoerythrin
- Peptide vaccine
- QOL, quality of life
- TGF-β, transforming growth factor-βl
- TNF, tumor necrosis factor
- Treg, regulatory T cell
- mAb, monoclonal antibody
Collapse
Affiliation(s)
- Norihiro Fujinami
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| | - Toshiaki Yoshikawa
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Yu Sawada
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Manami Shimomura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Tatsuaki Iwama
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Shiori Sugai
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| | - Shigehisa Kitano
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji, Tokyo, Japan
| | - Yasushi Uemura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| |
Collapse
|
6
|
Preclinical targeting of human T-cell malignancies using CD4-specific chimeric antigen receptor (CAR)-engineered T cells. Leukemia 2015; 30:701-7. [PMID: 26526988 DOI: 10.1038/leu.2015.311] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/26/2015] [Indexed: 12/21/2022]
Abstract
Peripheral T-cell lymphomas (PTCLs) are aggressive lymphomas with no effective upfront standard treatment and ineffective options in relapsed disease, resulting in poorer clinical outcomes as compared with B-cell lymphomas. The adoptive transfer of T cells engineered to express chimeric antigen receptors (CARs) is a promising new approach for treatment of hematological malignancies. However, preclinical reports of targeting T-cell lymphoma with CARs are almost non-existent. Here we have designed a CAR, CD4CAR, which redirects the antigen specificity of CD8+ cytotoxic T cells to CD4-expressing cells. CD4CAR T cells derived from human peripheral blood mononuclear cells and cord blood effectively redirected T-cell specificity against CD4+ cells in vitro. CD4CAR T cells efficiently eliminated a CD4+ leukemic cell line and primary CD4+ PTCL patient samples in co-culture assays. Notably, CD4CAR T cells maintained a central memory stem cell-like phenotype (CD8+CD45RO+CD62L+) under standard culture conditions. Furthermore, in aggressive orthotropic T-cell lymphoma models, CD4CAR T cells efficiently suppressed the growth of lymphoma cells while also significantly prolonging mouse survival. Combined, these studies demonstrate that CD4CAR-expressing CD8+ T cells are efficacious in ablating malignant CD4+ populations, with potential use as a bridge to transplant or stand-alone therapy for the treatment of PTCLs.
Collapse
|
7
|
Alzuguren P, Hervas-Stubbs S, Gonzalez-Aseguinolaza G, Poutou J, Fortes P, Mancheno U, Bunuales M, Olagüe C, Razquin N, Van Rooijen N, Enguita M, Hernandez-Alcoceba R. Transient depletion of specific immune cell populations to improve adenovirus-mediated transgene expression in the liver. Liver Int 2015; 35:1274-89. [PMID: 24754307 DOI: 10.1111/liv.12571] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 04/17/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Adenoviral (Ad) vectors are currently one of the most efficient tools for in vivo gene transfer to the liver. However, anti-Ad immune responses limit the safety and efficacy of these vectors. The initial inflammatory reaction is a concern in terms of toxicity, and it favours the development of cellular and humoral responses leading to short transgene persistence and inefficient vector re-administrations. Therefore, safe and simple ways to interfere with these processes are needed. Study ways to deplete specific immune cell populations and their impact on liver-directed gene transfer. METHODS First-generation Ad vectors encoding reporter genes (luciferase or β-galactosidase) were injected intravenously into Balb/c mice. Kupffer cells and splenic macrophages were depleted by intravenous administration of clodronate liposomes. B lymphocytes, CD4(+) , CD8(+) T lymphocytes or NK cells were depleted by intraperitoneal injection of anti-M plus anti-D, anti-CD4, anti-CD8 or anti-asialo-GM1 antibodies respectively. Long-term evolution of luciferase expression in the liver was monitored by bioluminescence imaging. RESULTS The anti-CD4 monoclonal antibody impaired cellular and humoral immune responses, leading to efficient vector re-administration. Clodronate liposomes had no impact on humoral responses but caused a 100-1000 fold increase in liver transduction, stabilized transgene expression, reduced the concentration of inflammatory cytokines, and inhibited lymphocyte activation. CONCLUSIONS Transient CD4(+) T-cell depletion using antibodies is a clinically feasible procedure that allows efficient Ad redosing. Systemic administration of clodronate liposomes may further increase the safety and efficacy of vectors.
Collapse
Affiliation(s)
- Pilar Alzuguren
- Division of Hepatology and Gene Therapy, CIMA, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Ueha S, Yokochi S, Ishiwata Y, Ogiwara H, Chand K, Nakajima T, Hachiga K, Shichino S, Terashima Y, Toda E, Shand FHW, Kakimi K, Ito S, Matsushima K. Robust Antitumor Effects of Combined Anti-CD4-Depleting Antibody and Anti-PD-1/PD-L1 Immune Checkpoint Antibody Treatment in Mice. Cancer Immunol Res 2015; 3:631-40. [PMID: 25711759 DOI: 10.1158/2326-6066.cir-14-0190] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/15/2015] [Indexed: 11/16/2022]
Abstract
Depletion of CD4(+) cells in tumor-bearing mice has strong antitumor effects. However, the mechanisms underlying these effects and the therapeutic benefits of CD4(+) cell depletion relative to other immunotherapies have not been fully evaluated. Here, we investigated the antitumor effects of an anti-CD4-depleting mAb as a monotherapy or in combination with immune checkpoint mAbs. In B16F10, Colon 26, or Lewis lung carcinoma subcutaneous tumor models, administration of the anti-CD4 mAb alone had strong antitumor effects that were superior to those elicited by CD25(+) Treg depletion or other immune checkpoint mAbs, and which were completely reversed by CD8(+) cell depletion. CD4(+) cell depletion led to the proliferation of tumor-specific CD8(+) T cells in the draining lymph node and increased infiltration of PD-1(+)CD8(+) T cells into the tumor, with a shift toward type I immunity within the tumor. Combination treatment with the anti-CD4 mAb and immune checkpoint mAbs, particularly anti-PD-1 or anti-PD-L1 mAbs, synergistically suppressed tumor growth and greatly prolonged survival. To our knowledge, this work represents the first report of robust synergy between anti-CD4 and anti-PD-1 or anti-PD-L1 mAb therapies.
Collapse
Affiliation(s)
- Satoshi Ueha
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shoji Yokochi
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. IDAC Theranostics, Inc., Tokyo, Japan
| | - Yoshiro Ishiwata
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. IDAC Theranostics, Inc., Tokyo, Japan
| | - Haru Ogiwara
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Krishant Chand
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takuya Nakajima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kosuke Hachiga
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. IDAC Theranostics, Inc., Tokyo, Japan
| | - Shigeyuki Shichino
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuya Terashima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Etsuko Toda
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Francis H W Shand
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Satoru Ito
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. IDAC Theranostics, Inc., Tokyo, Japan
| | - Kouji Matsushima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
9
|
|
10
|
Martin A, Tisch RM, Getts DR. Manipulating T cell-mediated pathology: Targets and functions of monoclonal antibody immunotherapy. Clin Immunol 2013; 148:136-47. [DOI: 10.1016/j.clim.2013.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 04/17/2013] [Accepted: 04/18/2013] [Indexed: 12/16/2022]
|
11
|
Elvin JG, Couston RG, van der Walle CF. Therapeutic antibodies: Market considerations, disease targets and bioprocessing. Int J Pharm 2013; 440:83-98. [DOI: 10.1016/j.ijpharm.2011.12.039] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 12/06/2011] [Accepted: 12/22/2011] [Indexed: 01/01/2023]
|
12
|
Izu-Belloso R, García-Ruiz J. Actualización terapéutica en linfomas cutáneos. ACTAS DERMO-SIFILIOGRAFICAS 2012; 103:694-707. [DOI: 10.1016/j.ad.2012.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 01/12/2012] [Accepted: 01/29/2012] [Indexed: 10/28/2022] Open
|
13
|
Izu-Belloso R, García-Ruiz J. Treatment of Cutaneous Lymphomas: an Update. ACTAS DERMO-SIFILIOGRAFICAS 2012. [DOI: 10.1016/j.adengl.2012.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
14
|
Abstract
Treatment regimens of patients with CTCL vary widely based on clinician preference and patient tolerance. Skin directed therapies are recommended for patients with early stage IA and IB MF, with combinations used in refractory cases. While no regimen has been proven to prolong survival in advanced stages, immunomodulatory regimens should be used initially to reduce the need for cytotoxic therapies. In more advanced stages of disease, treatment efforts should strive for palliation and improvement of quality of life. With many new therapies and strategies on the horizon, the future looks promising for CTCL patients. Unfortunately, other than allogeneic HCT, there are no potential curative therapies for CTCL. Clinical trials are currently underway to identify new therapies to improve quality of life for patients, and researchers are hard at work to identify novel pathways and genes for prognostication and as targets for therapies. Importantly, collaborative clinical trials to enhance rates of accrual need to be conducted, and improved interpretation of data via standardizing end points and response criteria should be an emphasis. Recently, the International Society for Cutaneous Lymphomas (ISCL), the United States Cutaneous Lymphoma Consortium (USCLC), and the Cutaneous Lymphoma Task Force of the European Organisation for Research and Treatment of Cancer (EORTC) met to develop consensus guidelines to facilitate collaboration on clinical trials. These proposed guidelines consist of: recommendations for standardizing general protocol design; a scoring system for assessing tumor burden in skin, lymph nodes, blood, and viscera; definition of response in skin, nodes, blood, and viscera; a composite global response score; and a definition of end points. Although these guidelines were generated by consensus panels, they have not been prospectively or retrospectively validated through analysis of large patient cohorts.
Collapse
|
15
|
Clark RA, Watanabe R, Teague JE, Schlapbach C, Tawa MC, Adams N, Dorosario AA, Chaney KS, Cutler CS, Leboeuf NR, Carter JB, Fisher DC, Kupper TS. Skin effector memory T cells do not recirculate and provide immune protection in alemtuzumab-treated CTCL patients. Sci Transl Med 2012; 4:117ra7. [PMID: 22261031 DOI: 10.1126/scitranslmed.3003008] [Citation(s) in RCA: 285] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cutaneous T cell lymphoma (CTCL) is a cancer of skin-homing T cells with variants that include leukemic CTCL (L-CTCL), a malignancy of central memory T cells (T(CM)), and mycosis fungoides (MF), a malignancy of skin resident effector memory T cells (T(EM)). We report that low-dose alemtuzumab (αCD52) effectively treated patients with refractory L-CTCL but not MF. Alemtuzumab depleted all T cells in blood and depleted both benign and malignant T(CM) from skin, but a diverse population of skin resident T(EM) remained in skin after therapy. T cell depletion with alemtuzumab required the presence of neutrophils, a cell type frequent in blood but rare in normal skin. These data suggest that T(CM) were depleted because they recirculate between the blood and the skin, whereas skin resident T(EM) were spared because they are sessile and non-recirculating. After alemtuzumab treatment, skin T cells produced lower amounts of interleukin-4 and higher amounts of interferon-γ. Moreover, there was a marked lack of infections in alemtuzumab-treated L-CTCL patients despite the complete absence of T cells in the blood, suggesting that skin resident T(EM) can protect the skin from pathogens even in the absence of T cell recruitment from the circulation. Together, these data suggest that alemtuzumab may treat refractory L-CTCL without severely compromising the immune response to infection by depleting circulating T(CM) but sparing the skin resident T(EM) that provide local immune protection of the skin.
Collapse
Affiliation(s)
- Rachael A Clark
- Department of Dermatology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Li JY, Horwitz S, Moskowitz A, Myskowski PL, Pulitzer M, Querfeld C. Management of cutaneous T cell lymphoma: new and emerging targets and treatment options. Cancer Manag Res 2012; 4:75-89. [PMID: 22457602 PMCID: PMC3308634 DOI: 10.2147/cmar.s9660] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cutaneous T cell lymphomas (CTCL) clinically and biologically represent a heterogeneous group of non-Hodgkin lymphomas, with mycosis fungoides and Sézary syndrome being the most common subtypes. Over the last decade, new immunological and molecular pathways have been identified that not only influence CTCL phenotype and growth, but also provide targets for therapies and prognostication. This review will focus on recent advances in the development of therapeutic agents, including bortezomib, the histone deacetylase inhibitors (vorinostat and romidepsin), and pralatrexate in CTCL.
Collapse
Affiliation(s)
- Janet Y Li
- College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | | | | | | | | | | |
Collapse
|
17
|
Wong HK, Mishra A, Hake T, Porcu P. Evolving insights in the pathogenesis and therapy of cutaneous T-cell lymphoma (mycosis fungoides and Sezary syndrome). Br J Haematol 2011; 155:150-66. [PMID: 21883142 DOI: 10.1111/j.1365-2141.2011.08852.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cutaneous T-cell lymphomas (CTCL) are a heterogeneous group of malignancies derived from skin-homing T cells. The most common forms of CTCL are Mycosis Fungoides (MF) and Sezary Syndrome (SS). Accurate diagnosis remains a challenge due to the heterogeneity of presentation and the lack of highly characteristic immunophenotypical and genetic markers. Over the past decade molecular studies have improved our understanding of the biology of CTCL. The identification of gene expression differences between normal and malignant T-cells has led to promising new diagnostic and prognostic biomarkers that now need validation to be incorporated into clinical practice. These biomarkers may also provide insight into the mechanism of development of CTCL. Additionally, treatment options have expanded with the approval of new agents, such as histone deacetylase inhibitors. A better understanding of the cell biology, immunology and genetics underlying the development and progression of CTCL will allow the design of more rational treatment strategies for these malignancies. This review summarizes the clinical epidemiology, staging and natural history of MF and SS; discusses the immunopathogenesis of MF and the functional role of the malignant T-cells; and reviews the latest advances in MF and SS treatment.
Collapse
Affiliation(s)
- Henry K Wong
- Division of Dermatology, The Ohio State University, Columbus, OH 43221, USA.
| | | | | | | |
Collapse
|
18
|
Alexandroff AB, Shpadaruk V, Bamford WM, Kennedy DBJ, Burd R, Dyer MJS. Alemtuzumab-resistant Sézary syndrome responding to zanolimumab. Br J Haematol 2011; 154:419-21. [PMID: 21480857 DOI: 10.1111/j.1365-2141.2011.08620.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
19
|
Abstract
INTRODUCTION The advent of anti-CD20 monoclonal antibody (mAb) rituximab heralded a new era in the treatment of non-Hodgkin's lymphoma leading to significant improvements in outcome for patients. This unprecedented success has changed the mindset of the clinical community and catalyzed the interest in the pharmaceutical industry to develop the next-generation of antibodies and antibody conjugates in cancer. AREAS COVERED There are an ever increasing number of newer generation anti-CD20 and rituximab 'bio-similars' undergoing early phase clinical development. In addition emerging novel therapies including antibody drug conjugates (brentuximab vedotin, SGN-35) and mAb against T-cell lymphomas antigens (e.g., zanolimumab) offer hope of improved outcome for other lymphomas. Bispecific T-cell-engaging antibodies and combination immunotherapy, also provide the promise of further improvements. Radiolabelled antibodies or radioimmunotherapy (RIT) has also demonstrated high clinical activity and two drugs namely 131I-tositumomab (Bexxar) and 90Y-ibritumomab (Zevalin) are licensed. EXPERT OPINION Despite the large numbers of new anti-CD20 mAb currently undergoing clinical testing, improving on clinical efficacy of rituximab is a substantial challenge. Further improvements in outcome for patients will require rigorous testing in well designed clinical trials alongside the translation of new insights into mechanism of mAb action that lead to improvements in clinical efficacy.
Collapse
Affiliation(s)
- Sam Mayes
- University of Manchester, Manchester Academic Health Science Centre, School of Cancer and Enabling Sciences, School of Medicine, Manchester, M20 4BX, UK
| | | | | |
Collapse
|
20
|
Abstract
This overview of 25 monoclonal antibody (mAb) and 5 Fc fusion protein therapeutics provides brief descriptions of the candidates, recently published clinical study results and on-going Phase 3 studies. In alphanumeric order, the 2011 therapeutic antibodies to watch list comprises AIN-457, bapineuzumab, brentuximab vedotin, briakinumab, dalotuzumab, epratuzumab, farletuzumab, girentuximab (WX-G250), naptumomab estafenatox, necitumumab, obinutuzumab, otelixizumab, pagibaximab, pertuzumab, ramucirumab, REGN88, reslizumab, solanezumab, T1h , teplizumab, trastuzumab emtansine, tremelimumab, vedolizumab, zalutumumab and zanolimumab. In alphanumeric order, the 2011 Fc fusion protein therapeutics to watch list comprises aflibercept, AMG-386, atacicept, Factor VIII and Factor IX-Fc. Commercially-sponsored mAb and Fc fusion therapeutics that have progressed only as far as Phase 2/3 or 3 were included. Candidates undergoing regulatory review or products that have been approved may also be in Phase 3 studies, but these were excluded. Due to the large body of primary literature about the candidates, only selected references are given and results from recent publications and articles that were relevant to Phase 3 studies are emphasized. Current as of September 2010, the information presented here will serve as a baseline against which future progress in the development of antibody-based therapeutics can be measured.
Collapse
|
21
|
D’Amore F, Radford J, Relander T, Jerkeman M, Tilly H, Österborg A, Morschhauser F, Gramatzki M, Dreyling M, Bang B, Hagberg H. Phase II trial of zanolimumab (HuMax-CD4) in relapsed or refractory non-cutaneous peripheral T cell lymphoma. Br J Haematol 2010; 150:565-73. [DOI: 10.1111/j.1365-2141.2010.08298.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
22
|
Mashima R, Hishida Y, Tezuka T, Yamanashi Y. The roles of Dok family adapters in immunoreceptor signaling. Immunol Rev 2010; 232:273-85. [PMID: 19909370 DOI: 10.1111/j.1600-065x.2009.00844.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The mammalian Dok protein family has seven members (Dok-1-Dok-7). The Dok proteins share structural similarities characterized by the NH2-terminal pleckstrin homology and phosphotyrosine-binding domains followed by SH2 target motifs in the COOH-terminal moiety, indicating an adapter function. Indeed, Dok-1 was originally identified as a 62 kDa protein that binds with p120 rasGAP, a potent inhibitor of Ras, upon tyrosine phosphorylation by a variety of protein tyrosine kinases. Among the Dok family, only Dok-1, Dok-2, and Dok-3 are preferentially expressed in hematopoietic/immune cells. Dok-1 and its closest relative Dok-2 act as negative regulators of the Ras-Erk pathway downstream of many immunoreceptor-mediated signaling systems, and it is believed that recruitment of p120 rasGAP by Dok-1 and Dok-2 is critical to their negative regulation. By contrast, Dok-3 does not bind with p120 rasGAP. However, accumulating evidence has demonstrated that Dok-3 is a negative regulator of the activation of JNK and mobilization of Ca2+ in B-cell receptor-mediated signaling, where the interaction of Dok-3 with SHIP-1 and Grb2 appears to be important. Here, we review the physiological roles and underlying mechanisms of Dok family proteins.
Collapse
Affiliation(s)
- Ryuichi Mashima
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
23
|
Erter J, Alinari L, Darabi K, Gurcan M, Garzon R, Marcucci G, Bechtel MA, Wong H, Porcu P. New targets of therapy in T-cell lymphomas. Curr Drug Targets 2010; 11:482-93. [PMID: 20196721 DOI: 10.2174/138945010790980376] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Accepted: 10/09/2009] [Indexed: 12/23/2022]
Abstract
T-cell lymphomas (TCL) are characterized by poor response to chemotherapy and generally poor outcome. While molecular profiling has identified distinct biological subsets and therapeutic targets in B-cell lymphomas, the molecular characterization of TCL has been slower. Surface markers expressed on malignant T-cells, such as CD2, CD3, CD4, CD25, and CD52 were the first TCL-specific therapeutic targets to be discovered. However, the presence of these receptors on normal T-cells means that monoclonal antibody (mAb)- or immunotoxin (IT)-based therapy in TCL inevitably results in variable degrees of immunosuppression. Thus, although some mAbs/IT have significant activity in selected subsets of TCL, more specific agents that target signaling pathways preferentially activated in malignant T-cells are needed. One such novel class of agents is represented by the histone deacetylase (HDAC) inhibitors. These molecules selectively induce apoptosis in a variety of transformed cells, including malignant T-cells, both in vitro and in vivo. Several HDAC inhibitors have been studied in TCL with promising results, and have recently been approved for clinical use. Immunomodulatory drugs, such as interferons and Toll Receptor (TLR) agonists have significant clinical activity in TCL, and are particularly important in the treatment of primary cutaneous subtypes (CTCL). Although most classical cytotoxic drugs have limited efficacy against TCL, agents that inhibit purine and pyrimidine metabolism, known as nucleoside analogues, and novel antifolate drugs, such as pralatrexate, are highly active in TCL. With improved molecular profiling of TCL novel pharmacological agents with activity in TCL are now being discovered at an increasingly rapid pace. Clinical trials are in progress and these agents are being integrated in combination therapies for TCL, both in the relapsed/refractory setting as well as front line.
Collapse
Affiliation(s)
- Jack Erter
- Division of Hematology - Oncology, The Ohio State University, Comprehensive Cancer Center, B-320 Starling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43210, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Steward-Tharp SM, Song YJ, Siegel RM, O'Shea JJ. New insights into T cell biology and T cell-directed therapy for autoimmunity, inflammation, and immunosuppression. Ann N Y Acad Sci 2010; 1183:123-48. [PMID: 20146712 PMCID: PMC2950114 DOI: 10.1111/j.1749-6632.2009.05124.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
T cell-directed therapies have become mainstays in the management of various autoimmune diseases and organ transplantation. The understanding of T cell biology has expanded greatly since the development of most agents currently in use. Here we discuss important recent discoveries pertaining to T helper cell differentiation, lineage commitment, and function. Within this context, we examine existing T cell-directed therapies, including new agents being evaluated in clinical and preclinical studies. We also use recent findings to speculate on novel targets.
Collapse
Affiliation(s)
- Scott M Steward-Tharp
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | |
Collapse
|
25
|
LaCelle MG, Jensen SM, Fox BA. Partial CD4 depletion reduces regulatory T cells induced by multiple vaccinations and restores therapeutic efficacy. Clin Cancer Res 2009; 15:6881-90. [PMID: 19903784 DOI: 10.1158/1078-0432.ccr-09-1113] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE A single vaccination of intact or reconstituted-lymphopenic mice (RLM) with a granulocyte macrophage colony-stimulating factor-secreting B16BL6-D5 melanoma cell line induces protective antitumor immunity and T cells that mediate the regression of established melanoma in adoptive immunotherapy studies. We wanted to study if multiple vaccinations during immune reconstitution of the lymphopenic host would maintain a potent antitumor immune response. EXPERIMENTAL DESIGN RLM were vaccinated multiple times over a 40-day period. Spleens were isolated from these mice, activated in vitro, and adoptively transferred into mice bearing 3-day experimental pulmonary metastases. RESULTS Multiple vaccinations, rather than boosting the immune response, significantly reduced therapeutic efficacy of adoptive immunotherapy and were associated with an increased frequency and absolute number of CD3+CD4+Foxp3+ T regulatory (T(reg)) cells. Anti-CD4 administration reduced the absolute number of T(reg) cells 9-fold. Effector T-cells generated from anti-CD4-treated mice were significantly (P < 0.0001) more therapeutic in adoptive transfer studies than T cells from multiply vaccinated animals with a full complement of CD4+ cells. CONCLUSION These results suggest that CD4+ T(reg) cells limit the efficacy of multiple vaccinations and that timed partial depletion of CD4+ T cells may reduce suppression and "tip-the-balance" in favor of therapeutic antitumor immunity. The recent failure of large phase III cancer vaccine clinical trials, wherein patients received multiple vaccines, underscores the potential clinical relevance of these findings.
Collapse
Affiliation(s)
- Michael G LaCelle
- Laboratory of Molecular and Tumor Immunology, Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Oregon Health and Science University, Portland, Oregon, USA
| | | | | |
Collapse
|
26
|
Abstract
Over the past decade, monoclonal antibodies have dramatically impacted the treatment of haematological malignancies, as evidenced by the effect of rituximab on the response rate and survival of patients with follicular and diffuse large B cell non-Hodgkin's lymphoma. Currently, only two monoclonal antibodies - the anti-CD33 immunotoxin gemtuzumab ozogamicin and the CD52-directed antibody alemtuzumab - are approved for treatment of relapsed acute myeloid leukaemia in older patients and B cell chronic lymphocytic leukaemia, respectively. Although not approved for such treatment, alemtuzumab is also active against T cell prolymphocytic leukaemia, cutaneous T cell lymphoma and Sézary syndrome, and adult T cell leukaemia and lymphoma. In addition, rituximab has demonstrated activity against B cell chronic lymphocytic and hairy cell leukaemia. Monoclonal antibodies targeting CD4, CD19, CD20, CD22, CD23, CD25, CD45, CD66 and CD122 are now being studied in the clinic for the treatment of leukaemia. Here, we discuss how these new antibodies have been engineered to reduce immunogenicity and improve antibody targeting and binding. Improved interactions with Fc receptors on immune effector cells can enhance destruction of target cells through antibody-dependent cellular cytotoxicity and complement-mediated cell lysis. The antibodies can also be armed with cellular toxins or radionuclides to enhance the destruction of leukaemia cells.
Collapse
Affiliation(s)
- John C Morris
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-1457, USA.
| | | |
Collapse
|
27
|
|
28
|
Ruuls SR, Lammerts van Bueren JJ, van de Winkel JGJ, Parren PWHI. Novel human antibody therapeutics: the age of the Umabs. Biotechnol J 2009; 3:1157-71. [PMID: 18702090 PMCID: PMC2959493 DOI: 10.1002/biot.200800110] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Monoclonal antibodies represent a major and increasingly important category of biotechnology products for the treatment of human diseases. The state-of-the-art of antibody technology has evolved to the point where therapeutic monoclonal antibodies, that are practically indistinguishable from antibodies induced in humans, are routinely generated. We depict how our science-based approach can be used to further improve the efficacy of antibody therapeutics, illustrated by the development of three monoclonal antibodies for various cancer indications: zanolimumab (directed against CD4), ofatumumab (directed against CD20) and zalutumumab (directed against epidermal growth factor receptor).
Collapse
|
29
|
Goodman OB, Dang NH. Novel antibody approaches for T-cell lymphomas. ACTA ACUST UNITED AC 2008; 8 Suppl 5:S193-8. [PMID: 19073527 DOI: 10.3816/clm.2008.s.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
T-cell lymphomas are biologically more aggressive--and less responsive than their B-cell counterparts--to conventional chemotherapy. The understanding of the normal physiology of T-cell signaling as well as the pathophysiology of T-cell neoplasia has advanced dramatically in the past decade. Thus, a number of novel cell surface therapeutic targets specific for T-cells have been identified and evaluated as potential monoclonal antibody (MoAb)-based therapeutic targets. Herein, we review a number of these cell surface targets and discuss the development and the clinical evaluation of MoAb therapies directed against these antigens.
Collapse
Affiliation(s)
- Oscar B Goodman
- Department of Clinical Oncology, Nevada Cancer Institute, Las Vegas, NV 89135, USA
| | | |
Collapse
|
30
|
Sebastian Mestel D, Beyer M, Möbs M, Steinhoff M, Sterry W, Assaf C. Zanolimumab, a human monoclonal antibody targeting CD4 in the treatment of mycosis fungoides and Sézary syndrome. Expert Opin Biol Ther 2008; 8:1929-39. [DOI: 10.1517/14712590802528696] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
31
|
Agostinelli C, Piccaluga PP, Went P, Rossi M, Gazzola A, Righi S, Sista T, Campidelli C, Zinzani PL, Falini B, Pileri SA. Peripheral T cell lymphoma, not otherwise specified: the stuff of genes, dreams and therapies. J Clin Pathol 2008; 61:1160-7. [PMID: 18755717 PMCID: PMC2582342 DOI: 10.1136/jcp.2008.055335] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Peripheral T cell lymphomas (PTCL) account for about 12% of lymphoid tumours worldwide. Almost half show such morphological and molecular variability as to hamper any further classification, and to justify their inclusion in a waste-basket category termed “not otherwise specified (NOS)”. The latter term is used for neoplasms with aggressive presentation, poor response to therapy and dismal prognosis. In contrast to B cell lymphomas, PTCL have been the subject of only a limited number of studies to elucidate their pathobiology and identify novel pharmacological approaches. Herewith, the authors revise the most recent contributions on the subject based on the experience they have gained in the extensive application of microarray technologies. PTCL/NOS are characterised by erratic expression of T cell associated antigens, including CD4 and CD52, which have recently been proposed as targets for ad hoc immunotherapies. PTCL/NOS also show variable Ki-67 marking, with rates >80% heralding a worse prognosis. Gene expression profiling studies have revealed that PTCL/NOS derive from activated T lymphocytes, more often of the CD4+ type, and bear a signature composed of 155 genes and related products that play a pivotal role in cell signalling transduction, proliferation, apoptosis and matrix remodelling. This observation seems to pave the way for the use of innovative drugs such as tyrosine kinase and histone deacetylase inhibitors whose efficacy has been proven in PTCL primary cell cultures. Gene expression profiling also allows better distinction of PTCL/NOS from angioimmunoblastic T cell lymphoma, the latter being characterised by follicular T helper lymphocyte derivation and CXCL13, PD1 and vascular endothelial growth factor expression.
Collapse
Affiliation(s)
- C Agostinelli
- Department of Haematology and Clinical Oncology L and A Seràgnoli, Bologna University School of Medicine, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|