Nanobody-derived bispecific CAR-T cell therapy enhances the anti-tumor efficacy of T cell lymphoma treatment
暂无分享,去创建一个
Xin He | Yingtong Lin | Rong Li | Fan Zou | Bingfeng Liu | Hui Zhang | Wenyu Li | Yidan Qiao | Keming Lin | Cancan Chen | Mo Zhou | Baijin Xia | Yuzhuang Li | Wanying Zhang | Fei-li Chen | Linghua Li | Lijuan Lu | Xuemei Wang
[1] Shuhong Li,et al. A novel CD19/CD22/CD3 trispecific antibody enhances therapeutic efficacy and overcomes immune escape against B-ALL. , 2022, Blood.
[2] Zhaoming Li,et al. Autologous nanobody-derived fratricide-resistant CD7-CAR T cell therapy for patients with relapsed and refractory T-cell acute lymphoblastic leukemia/lymphoma. , 2022, Clinical cancer research : an official journal of the American Association for Cancer Research.
[3] Jiali Cheng,et al. T cells expressing CD5/CD7 bispecific chimeric antigen receptors with fully human heavy-chain-only domains mitigate tumor antigen escape , 2022, Signal Transduction and Targeted Therapy.
[4] Ansuman T. Satpathy,et al. Surface proteomics reveals CD72 as a target for in vitro-evolved nanobody-based CAR-T cells in KMT2A/MLL1-rearranged B-ALL. , 2021, Cancer discovery.
[5] D. Chihara,et al. Targeted based therapy in nodal T-cell lymphomas , 2021, Leukemia.
[6] Weiqi Wang,et al. Nanobody-based chimeric antigen receptor T cells designed by CRISPR/Cas9 technology for solid tumor immunotherapy , 2021, Signal Transduction and Targeted Therapy.
[7] Jun Zhao,et al. Nanobody: a promising toolkit for molecular imaging and disease therapy , 2021, EJNMMI Research.
[8] G. Przybylski,et al. Novel targeted therapies of T cell lymphomas , 2020, Journal of Hematology & Oncology.
[9] H. Ploegh,et al. Nanobodies in cancer. , 2020, Seminars in immunology.
[10] Zhiqiang Wu,et al. Optimized tandem CD19/CD20 CAR-engineered T cells in refractory/relapsed B cell lymphoma. , 2020, Blood.
[11] W. Chan,et al. Peripheral T cell lymphomas: from the bench to the clinic , 2020, Nature Reviews Cancer.
[12] H. Ploegh,et al. Improved Antitumor Efficacy of Chimeric Antigen Receptor T Cells that Secrete Single-Domain Antibody Fragments , 2020, Cancer Immunology Research.
[13] J. Tavernier,et al. Rapid and Effective Generation of Nanobody Based CARs using PCR and Gibson Assembly , 2020, International journal of molecular sciences.
[14] Meijuan Huang,et al. Engineered triple inhibitory receptor resistance improves anti-tumor CAR-T cell performance via CD56 , 2019, Nature Communications.
[15] Zhiqiang Wu,et al. Haploidentical CD19/CD22 bispecific CAR-T cells induced MRD-negative remission in a patient with relapsed and refractory adult B-ALL after haploidentical hematopoietic stem cell transplantation , 2019, Journal of Hematology & Oncology.
[16] R. Hynes,et al. Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice , 2019, Proceedings of the National Academy of Sciences.
[17] P. Dreger. Comment to “The outcome of peripheral T-cell lymphoma patients failing first-line therapy” , 2019, Haematologica.
[18] U. Rix,et al. An immunoproteomic approach to characterize the CAR interactome and signalosome , 2019, Science Signaling.
[19] J-Pablo Salvador,et al. Nanobody: outstanding features for diagnostic and therapeutic applications , 2019, Analytical and Bioanalytical Chemistry.
[20] B. Lei,et al. A phase 1, open-label study of LCAR-B38M, a chimeric antigen receptor T cell therapy directed against B cell maturation antigen, in patients with relapsed or refractory multiple myeloma , 2018, Journal of Hematology & Oncology.
[21] Christopher A. Miller,et al. An ‘off-the-shelf’ fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies , 2018, Leukemia.
[22] N. Pemmaraju. Novel Pathways and Potential Therapeutic Strategies for Blastic Plasmacytoid Dendritic Cell Neoplasm (BPDCN): CD123 and Beyond , 2017, Current Hematologic Malignancy Reports.
[23] Hao Liu,et al. Clinical and immunological responses after CD30-specific chimeric antigen receptor-redirected lymphocytes. , 2017, The Journal of clinical investigation.
[24] T. Waldmann,et al. Chimeric antigen receptor modified T cells that target chemokine receptor CCR4 as a therapeutic modality for T‐cell malignancies , 2017, American journal of hematology.
[25] Ciaran M. Lee,et al. CD7-edited T cells expressing a CD7-specific CAR for the therapy of T-cell malignancies. , 2017, Blood.
[26] A. Kuhn,et al. The Transmembrane Morphogenesis Protein gp1 of Filamentous Phages Contains Walker A and Walker B Motifs Essential for Phage Assembly , 2017, Viruses.
[27] Michael L. Wang,et al. The survival outcome of patients with relapsed/refractory peripheral T‐cell lymphoma‐not otherwise specified and angioimmunoblastic T‐cell lymphoma , 2017, British journal of haematology.
[28] H. Tony,et al. Resistance to anti-CD19/CD3 BiTE in acute lymphoblastic leukemia may be mediated by disrupted CD19 membrane trafficking. , 2017, Blood.
[29] J. Cerhan,et al. 2016 US lymphoid malignancy statistics by World Health Organization subtypes , 2016, CA: a cancer journal for clinicians.
[30] S. Grupp,et al. Dual CD19 and CD123 targeting prevents antigen-loss relapses after CD19-directed immunotherapies. , 2016, The Journal of clinical investigation.
[31] Yang Liu,et al. Autologous T Cells Expressing CD30 Chimeric Antigen Receptors for Relapsed or Refractory Hodgkin Lymphoma: An Open-Label Phase I Trial , 2016, Clinical Cancer Research.
[32] Xu Zhang,et al. Chimeric Antigen Receptor T Cells Guided by the Single-Chain Fv of a Broadly Neutralizing Antibody Specifically and Effectively Eradicate Virus Reactivated from Latency in CD4+ T Lymphocytes Isolated from HIV-1-Infected Individuals Receiving Suppressive Combined Antiretroviral Therapy , 2016, Journal of Virology.
[33] M. Cazzola. Introduction to a review series: the 2016 revision of the WHO classification of tumors of hematopoietic and lymphoid tissues. , 2016, Blood.
[34] Y. Chen,et al. T Cells Expressing CD19/CD20 Bispecific Chimeric Antigen Receptors Prevent Antigen Escape by Malignant B Cells , 2016, Cancer Immunology Research.
[35] Yupo Ma,et al. Preclinical targeting of human T-cell malignancies using CD4-specific chimeric antigen receptor (CAR)-engineered T cells , 2016, Leukemia.
[36] M. Brenner,et al. A T-cell-directed chimeric antigen receptor for the selective treatment of T-cell malignancies. , 2015, Blood.
[37] K. Savage,et al. Belinostat in Patients With Relapsed or Refractory Peripheral T-Cell Lymphoma: Results of the Pivotal Phase II BELIEF (CLN-19) Study. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[38] S. Oliveira,et al. Nanobody-based cancer therapy of solid tumors. , 2015, Nanomedicine.
[39] Yang Liu,et al. Effective response and delayed toxicities of refractory advanced diffuse large B-cell lymphoma treated by CD20-directed chimeric antigen receptor-modified T cells. , 2014, Clinical immunology.
[40] Pamela A Shaw,et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. , 2014, The New England journal of medicine.
[41] M. Jerkeman,et al. Real-world data on prognostic factors and treatment in peripheral T-cell lymphomas: a study from the Swedish Lymphoma Registry. , 2014, Blood.
[42] H. Wajant,et al. ARGX-110, a highly potent antibody targeting CD70, eliminates tumors via both enhanced ADCC and immune checkpoint blockade , 2014, mAbs.
[43] J. Cortes,et al. CD30 expression in acute lymphoblastic leukemia as assessed by flow cytometry analysis , 2014, Leukemia & lymphoma.
[44] D. Weisenburger,et al. Differences in incidence and trends of haematological malignancies in Japan and the United States , 2013, British journal of haematology.
[45] S. Magez,et al. Novel therapy based on camelid nanobodies. , 2013, Therapeutic delivery.
[46] M. Kalos,et al. Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. , 2013, Immunity.
[47] M. Shokrgozar,et al. Genetically engineered T cells bearing chimeric nanoconstructed receptors harboring TAG-72-specific camelid single domain antibodies as targeting agents. , 2013, Cancer letters.
[48] R. Gascoyne,et al. Survival of patients with peripheral T-cell lymphoma after first relapse or progression: spectrum of disease and rare long-term survivors. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[49] Michel Sadelain,et al. The basic principles of chimeric antigen receptor design. , 2013, Cancer discovery.
[50] R. Pazdur,et al. U.S. Food and Drug Administration Approval Summary: Brentuximab Vedotin for the Treatment of Relapsed Hodgkin Lymphoma or Relapsed Systemic Anaplastic Large-Cell Lymphoma , 2012, Clinical Cancer Research.
[51] R. Advani,et al. Brentuximab vedotin (SGN-35) in patients with relapsed or refractory systemic anaplastic large-cell lymphoma: results of a phase II study. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[52] Matthew Greenwood,et al. Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T-cell lymphoma after prior systemic therapy. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[53] M. Dhodapkar,et al. Harnessing natural killer T (NKT) cells in human myeloma: progress and challenges. , 2011, Clinical immunology.
[54] K. Savage,et al. Pralatrexate in patients with relapsed or refractory peripheral T-cell lymphoma: results from the pivotal PROPEL study. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[55] W. Wilson,et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. , 2010, Blood.
[56] H. Heslop,et al. Epstein Barr virus specific cytotoxic T lymphocytes expressing the anti-CD30zeta artificial chimeric T-cell receptor for immunotherapy of Hodgkin disease. , 2007, Blood.
[57] Mark Ende,et al. Hematopoietic stem-cell transplantation. , 2006, The New England journal of medicine.
[58] S. Pileri,et al. Marker expression in peripheral T-cell lymphoma: a proposed clinical-pathologic prognostic score. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[59] Andrew Gibson,et al. AP2 Adaptor Complex-Dependent Internalization of CD5: Differential Regulation in T and B Cells1 , 2002, The Journal of Immunology.
[60] W. Leonard. Cytokines and immunodeficiency diseases , 2001, Nature Reviews Immunology.
[61] C Cambillau,et al. Thermal unfolding of a llama antibody fragment: a two-state reversible process. , 2001, Biochemistry.
[62] B. de Geus,et al. Llama heavy-chain V regions consist of at least four distinct subfamilies revealing novel sequence features. , 2000, Molecular immunology.
[63] J. Roberts,et al. Hematopoietic stem-cell transplantation for the treatment of severe combined immunodeficiency. , 1999, The New England journal of medicine.
[64] I. Pastan,et al. Isolation of a high-affinity stable single-chain Fv specific for mesothelin from DNA-immunized mice by phage display and construction of a recombinant immunotoxin with anti-tumor activity. , 1998, Proceedings of the National Academy of Sciences of the United States of America.
[65] L. Wyns,et al. Selection and identification of single domain antibody fragments from camel heavy‐chain antibodies , 1997, FEBS letters.
[66] G. Studnicka,et al. Human-engineered monoclonal antibodies retain full specific binding activity by preserving non-CDR complementarity-modulating residues. , 1994, Protein engineering.
[67] S. Muyldermans,et al. Naturally occurring antibodies devoid of light chains , 1993, Nature.
[68] Z. Eshhar,et al. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. , 1989, Proceedings of the National Academy of Sciences of the United States of America.
[69] Harris,et al. Introduction to a review series: the 2016 revision of the WHO classi fi cation of tumors of hematopoietic and lymphoid tissues , 2016 .
[70] F. Stirpe,et al. CD30 (Ki-1) molecule: a new cytokine receptor of the tumor necrosis factor receptor superfamily as a tool for diagnosis and immunotherapy. , 1995, Blood.