1
|
Abdel-Azim H, Dave H, Jordan K, Rawlings-Rhea S, Luong A, Wilson AL. Alignment of practices for data harmonization across multi-center cell therapy trials: a report from the Consortium for Pediatric Cellular Immunotherapy. Cytotherapy 2022; 24:193-204. [PMID: 34711500 PMCID: PMC8792313 DOI: 10.1016/j.jcyt.2021.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/13/2021] [Accepted: 08/27/2021] [Indexed: 02/03/2023]
Abstract
Immune effector cell (IEC) therapies have revolutionized our approach to relapsed B-cell malignancies, and interest in the investigational use of IECs is rapidly expanding into other diseases. Current challenges in the analysis of IEC therapies include small sample sizes, limited access to clinical trials and a paucity of predictive biomarkers of efficacy and toxicity associated with IEC therapies. Retrospective and prospective multi-center cell therapy trials can assist in overcoming these barriers through harmonization of clinical endpoints and correlative assays for immune monitoring, allowing additional cross-trial analysis to identify biomarkers of failure and success. The Consortium for Pediatric Cellular Immunotherapy (CPCI) offers a unique platform to address the aforementioned challenges by delivering cutting-edge cell and gene therapies for children through multi-center clinical trials. Here the authors discuss some of the important pre-analytic variables, such as biospecimen collection and initial processing procedures, that affect biomarker assays commonly used in IEC trials across participating CPCI sites. The authors review the recent literature and provide data to support recommendations for alignment and standardization of practices that can affect flow cytometry assays measuring immune effector function as well as interpretation of cytokine/chemokine data. The authors also identify critical gaps that often make parallel comparisons between trials difficult or impossible.
Collapse
Affiliation(s)
- Hisham Abdel-Azim
- Cancer and Blood Disease Institute, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Hema Dave
- Center for Cancer and Blood Disorders, Children's National Hospital, George Washington School of Medicine, Washington, DC, USA
| | - Kimberly Jordan
- Department of Immunology and Microbiology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Stephanie Rawlings-Rhea
- Seattle Children's Therapeutics, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Annie Luong
- Cancer and Blood Disease Institute, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ashley L Wilson
- Seattle Children's Therapeutics, Seattle Children's Research Institute, Seattle, Washington, USA.
| |
Collapse
|
2
|
Hashemzadeh N, Dolatkhah M, Adibkia K, Aghanejad A, Barzegar-Jalali M, Omidi Y, Barar J. Recent advances in breast cancer immunotherapy: The promising impact of nanomedicines. Life Sci 2021; 271:119110. [PMID: 33513401 DOI: 10.1016/j.lfs.2021.119110] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/29/2022]
Abstract
Breast cancer (BC) is one of the prevalent cancers among women. Generally, the treatment of BC is mostly based on several prominent strategies, including chemotherapy, surgery, endocrine therapy, molecular targeted therapy, and radiation. Owing to the growing knowledge about the complexity of BC pathobiology, immunotherapy as a promising treatment modality has substantially improved the patients' care in the clinic. Immunotherapy is used to harness the patient's immune system to recognize and battle devious cancer cells. As a novel therapy approach, this emerging strategy targets the key molecular entities of tumor tissue. To achieve maximal therapeutic impacts, the dynamic interplay between cancer and immune cells needs to be fully comprehended. The key molecular machinery of solid tumors can be targeted by nanoscale immunomedicines. While discussing the potential biomarkers involved in the initiation and progression of BC, we aimed to provide comprehensive insights into the immunotherapy and articulate the recent advances in terms of the therapeutic strategies used to control this disease, including immune checkpoint inhibitors, vaccines, chimeric antigen receptor T cells therapy, and nanomedicines.
Collapse
Affiliation(s)
- Nastaran Hashemzadeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mitra Dolatkhah
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khosro Adibkia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Barzegar-Jalali
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Nishimura T, Hsu I, Martinez-Krams DC, Nakauchi Y, Majeti R, Yamazaki S, Nakauchi H, Wilkinson AC. Use of polyvinyl alcohol for chimeric antigen receptor T-cell expansion. Exp Hematol 2019; 80:16-20. [PMID: 31874780 PMCID: PMC7194120 DOI: 10.1016/j.exphem.2019.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022]
Abstract
Serum albumin has long been an essential supplement for ex vivo hematopoietic and immune cell cultures. However, serum albumin medium supplements represent a major source of biological contamination in cell cultures and often cause loss of cellular function. As serum albumin exhibits significant batch-to-batch variability, it has also been blamed for causing major issues in experimental reproducibility. We recently discovered the synthetic polymer polyvinyl alcohol (PVA) as an inexpensive, Good Manufacturing Practice-compatible, and biologically inert serum albumin replacement for ex vivo hematopoietic stem cell cultures. Importantly, PVA is free of the biological contaminants that have plagued serum albumin-based media. Here, we describe that PVA can replace serum albumin in a range of blood and immune cell cultures including cell lines, primary leukemia samples, and human T lymphocytes. PVA can even replace human serum in the generation and expansion of functional chimeric antigen receptor (CAR) T cells, offering a potentially safer and more cost-efficient approach for this clinical cell therapy. In summary, PVA represents a chemically defined, biologically inert, and inexpensive alternative to serum albumin for a range of cell cultures in hematology and immunology.
Collapse
Affiliation(s)
- Toshinobu Nishimura
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA; Department of Genetics, Stanford University School of Medicine, Stanford, CA
| | - Ian Hsu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA; Department of Genetics, Stanford University School of Medicine, Stanford, CA
| | - Daniel C Martinez-Krams
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA; Department of Hematology, Stanford University School of Medicine, Stanford, CA
| | - Yusuke Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA; Department of Hematology, Stanford University School of Medicine, Stanford, CA
| | - Ravindra Majeti
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA; Department of Hematology, Stanford University School of Medicine, Stanford, CA
| | - Satoshi Yamazaki
- Division of Stem Cell Biology, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiromitsu Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA; Department of Genetics, Stanford University School of Medicine, Stanford, CA; Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
| | - Adam C Wilkinson
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA; Department of Genetics, Stanford University School of Medicine, Stanford, CA.
| |
Collapse
|
4
|
CD133-directed CAR T-cells for MLL leukemia: on-target, off-tumor myeloablative toxicity. Leukemia 2019; 33:2090-2125. [PMID: 30778134 PMCID: PMC6756031 DOI: 10.1038/s41375-019-0418-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/28/2018] [Accepted: 12/12/2018] [Indexed: 12/28/2022]
|
5
|
Jin Z, Xu L, Li Y. Approaches for generation of anti-leukemia specific T cells. CELL REGENERATION 2019; 7:40-44. [PMID: 30671229 PMCID: PMC6326242 DOI: 10.1016/j.cr.2018.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/13/2018] [Accepted: 09/26/2018] [Indexed: 02/06/2023]
Abstract
As three decades ago, it was reported that adoptive T cell immunotherapy by infusion of autologous tumor infiltrating lymphocytes (TILs) mediated objective cancer regression in patients with metastatic melanoma. A new era of T cell immunotherapy arose since the improvement and clinical use of anti-CD19 chimeric antigen receptor T cells (CAR-T) for the treatment of refractory and relapsed B lymphocyte leukemia. However, several challenges and difficulties remain on the way to reach generic and effective T cell immunotherapy, including lacking a generic method for generating anti-leukemia-specific T cells from every patient. Here, we summarize the current methods of generating anti-leukemia-specific T cells, and the promising approaches in the future.
Collapse
Key Words
- ACT, adoptive cellular immunotherapy
- APL, promyelocytic leukemia
- Anti-leukemia T cell
- B-ALL, cell acute lymphoblastic leukemia
- CAR-T
- CAR-T, chimeric antigen receptor T cells
- CML, chronic myelogenous leukemia
- CR, complete remission
- CTLs, cytotoxic T cells
- DLI, donor lymphocyte infusion
- FLT3-ITD, FLT3 internal tandem duplication
- GVHD, graft-versus-host disease
- GVL, graft-versus-leukemia
- HLA, human leukocyte antigen
- HPCs, hematopoietic progenitor cells
- IL-2, interleukin-2
- Ig, immunoglobulin
- T cell immunotherapy
- T cell reprogramming
- TAA, tumor-associated antigen
- TCR-T
- TCR-T, TCR gene-modified T cell
- TIL, infiltrating lymphocytes
- TKI, tyrosine kinase inhibitor
- WT1, Wilm's tumor antigen 1
- allo-HSCT, allogeneic hematopoietic stem cell transplantation
- hESC, human embryonic stem cell
- iPSCs, induced pluripotent stem cells
- iTs, induced functional T cells
- scFv, single-chain variable fragment
Collapse
Affiliation(s)
- Zhenyi Jin
- Key Laboratory for Regenerative Medicine of Ministry of Education; Institute of Hematology, School of Medicine; Jinan University, Guangzhou, 510632, China.,Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Ling Xu
- Key Laboratory for Regenerative Medicine of Ministry of Education; Institute of Hematology, School of Medicine; Jinan University, Guangzhou, 510632, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education; Institute of Hematology, School of Medicine; Jinan University, Guangzhou, 510632, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| |
Collapse
|