1
|
Mir M, Faiz S, Bommakanti AG, Sheshadri A. Pulmonary Immunocompromise in Stem Cell Transplantation and Cellular Therapy. Clin Chest Med 2025; 46:129-147. [PMID: 39890284 DOI: 10.1016/j.ccm.2024.10.010] [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] [Indexed: 02/03/2025]
Abstract
Hematopoietic cell transplantation (HCT) and cellular therapies, such as chimeric-antigen receptor T-cell (CAR-T) treatments, are potentially curative treatments for certain hematologic malignancies and some nonmalignant disorders. However, pulmonary complications, both infectious and noninfectious, remain a significant cause of morbidity and mortality in patients who receive cellular therapies. This review article provides an overview of pulmonary complications encountered in the context of HCT and CAR-T. The authors discuss mechanisms of underlying immunocompromise that lead to a rise in infections. Additionally, they highlight key noninfectious complications of HCT that can mimic acute infections and suggest diagnostic approaches and preventive strategies to distinguish these entities promptly.
Collapse
Affiliation(s)
- Mahnoor Mir
- Divisions of Critical Care, Pulmonary and Sleep Medicine, McGovern Medical School at UTHealth, Houston, TX 77030, USA; Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Saadia Faiz
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anuradha G Bommakanti
- Divisions of Critical Care, Pulmonary and Sleep Medicine, McGovern Medical School at UTHealth, Houston, TX 77030, USA; Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
2
|
Thirumalaisamy R, Vasuki S, Sindhu SM, Mothilal TM, Srimathi V, Poornima B, Bhuvaneswari M, Hariharan M. FDA-Approved Chimeric Antigen Receptor (CAR)-T Cell Therapy for Different Cancers-A Recent Perspective. Mol Biotechnol 2025; 67:469-483. [PMID: 38459361 DOI: 10.1007/s12033-024-01090-0] [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: 11/22/2023] [Accepted: 01/26/2024] [Indexed: 03/10/2024]
Abstract
Cancer is one of the most prevalent diseases in the world, and their rate of occurence has been increased in recent decades. Current review article, summarizes the novel treatment options Chimeric Antigen Receptor-T (CAR-T) cell therapy for various cancers constitute a major health and development challenge, impacting every aspect of sustainable development quoted by goal 3 good health and well-being of UN sustainable goals. WHO estimates that 70% of cancer deaths occur in low- and middle- income countries (LMICs) by 2030, LMICs are expected to bear the brunt of the expected 24.1 million new cancer cases per year. This current review article focuses and discussed about CAR-T cell therapy for various cancers against most prevalent non-communicable disease cancer disease stipulated by WHO and UN sustainable goals. Three literature databases Google scholar, Science Direct, PubMed was utilized to search and collect CAR-T cell treatment options for different cancers published articles sources in between January 2000 and December 2023. There were a total of 18,700 papers found, with 48 of them being found to be eligible focusing various cancer treatment by CAR-T cells utilized for the study. Based on the information gathered, CAR-T cell therapy treating different cancers and their merit and its advantages in heal and improve certain cancers was also discussed in this review article with their detailed molecular mechanisms. This article also gives an insight to utilize CAR-T cell treatment protocols for rejuvenating cancer patient from such ruthless cancer disease condition thereby improving life span of cancer patients and eradication of disease in some cases.
Collapse
Affiliation(s)
- R Thirumalaisamy
- Department of Biotechnology, Sona College Arts and Science, Salem (Dt.), Tamil Nadu, 636005, India.
| | - S Vasuki
- Department of Biotechnology, Sona College Arts and Science, Salem (Dt.), Tamil Nadu, 636005, India
| | - S M Sindhu
- Department of Biotechnology, Sona College Arts and Science, Salem (Dt.), Tamil Nadu, 636005, India
- Department of Biotechnology, PSGR Krishnammal College for Women (Autonomous), Coimbatore (Dt.), Tamil Nadu, 641004, India
| | - T M Mothilal
- Department of Biotechnology, Sona College Arts and Science, Salem (Dt.), Tamil Nadu, 636005, India
| | - V Srimathi
- Department of Biotechnology, Sona College Arts and Science, Salem (Dt.), Tamil Nadu, 636005, India
| | - B Poornima
- Department of Biotechnology, Sona College Arts and Science, Salem (Dt.), Tamil Nadu, 636005, India
| | - M Bhuvaneswari
- Department of Biotechnology, Sona College Arts and Science, Salem (Dt.), Tamil Nadu, 636005, India
| | - Mohan Hariharan
- Center for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamilnadu, 602105, India
| |
Collapse
|
3
|
Joseph T, Sanchez J, Abbasi A, Zhang L, Sica RA, Duong TQ. Cardiotoxic Effects Following CAR-T Cell Therapy: A Literature Review. Curr Oncol Rep 2025:10.1007/s11912-024-01634-2. [PMID: 39836349 DOI: 10.1007/s11912-024-01634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2024] [Indexed: 01/22/2025]
Abstract
PURPOSE OF REVIEW This paper reviewed the current literature on incidence, clinical manifestations, and risk factors of Chimeric Antigen Receptor T-cell (CAR-T) cardiotoxicity. RECENT FINDINGS CAR-T therapy has emerged as a groundbreaking treatment for hematological malignancies since FDA approval in 2017. CAR-T therapy is however associated with a few side effects, among which cardiotoxicity is of significant concern. There were only a few studies on CAR-T cardiotoxicity published to date with limited sample sizes, and their findings were heterogeneous. It was difficult to reach generalizable conclusions. CAR-T therapy was associated with significant risks for acute and subacute cardiotoxicity, as measured by echocardiograms, EKG, and blood biomarkers. Patients with cytokine release syndrome (CRS) grade 2 or higher were more likely to exhibit cardiotoxicity. The most prevalent cardiac events included hypotension-requiring inotropic or vasopressor support, tachycardia, heart failure/decompensation, atrial fibrillation, new or worsening cardiomyopathy, arrhythmia, myocarditis, cardiac arrest, and cardiovascular death. The most prevalent echocardiographic changes were systolic dysfunction and diastolic dysfunction, and abnormal echocardiogram findings. There were differences in findings between adult and pediatric patients. The long-term effects beyond a year post treatment remain largely unknown and long-term follow-up studies are warranted.
Collapse
Affiliation(s)
- Tony Joseph
- Department of Radiology, Albert Einstein College of Medicine and the Montefiore Medical Center, 111 East 210Th Street, Bronx, NY, 10461, USA
- Department of Chemistry, CUNY Brooklyn College, 2900 Bedford Ave, Brooklyn, NY, 11210, USA
| | - Jimmy Sanchez
- Department of Radiology, Albert Einstein College of Medicine and the Montefiore Medical Center, 111 East 210Th Street, Bronx, NY, 10461, USA
| | - Ahmed Abbasi
- Department of Oncology, Albert Einstein College of Medicine and the Montefiore Medical Center, 111 East 210Th Street, Bronx, NY, 10461, USA
| | - Lili Zhang
- Department of Medicine, Cardiology Division, Albert Einstein College of Medicine and the Montefiore Medical Center, 111 East 210Th Street, Bronx, NY, 10461, USA
| | - R Alejandro Sica
- Department of Oncology, Albert Einstein College of Medicine and the Montefiore Medical Center, 111 East 210Th Street, Bronx, NY, 10461, USA
| | - Tim Q Duong
- Department of Radiology, Albert Einstein College of Medicine and the Montefiore Medical Center, 111 East 210Th Street, Bronx, NY, 10461, USA.
| |
Collapse
|
4
|
Kong Y, Li J, Zhao X, Wu Y, Chen L. CAR-T cell therapy: developments, challenges and expanded applications from cancer to autoimmunity. Front Immunol 2025; 15:1519671. [PMID: 39850899 PMCID: PMC11754230 DOI: 10.3389/fimmu.2024.1519671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Chimeric Antigen Receptor (CAR)-T cell therapy has rapidly emerged as a groundbreaking approach in cancer treatment, particularly for hematologic malignancies. However, the application of CAR-T cell therapy in solid tumors remains challenging. This review summarized the development of CAR-T technologies, emphasized the challenges and solutions in CAR-T cell therapy for solid tumors. Also, key innovations were discussed including specialized CAR-T, combination therapies and the novel use of CAR-Treg, CAR-NK and CAR-M cells. Besides, CAR-based cell therapy have extended its reach beyond oncology to autoimmune disorders. We reviewed preclinical experiments and clinical trials involving CAR-T, Car-Treg and CAAR-T cell therapies in various autoimmune diseases. By highlighting these cutting-edge developments, this review underscores the transformative potential of CAR technologies in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Yanwei Wu
- School of Medicine, Shanghai University, Shanghai, China
| | - Liang Chen
- School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
5
|
Marei HE, Bedair K, Hasan A, Al-Mansoori L, Caratelli S, Sconocchia G, Gaiba A, Cenciarelli C. Current status and innovative developments of CAR-T-cell therapy for the treatment of breast cancer. Cancer Cell Int 2025; 25:3. [PMID: 39755633 DOI: 10.1186/s12935-024-03615-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/12/2024] [Indexed: 01/06/2025] Open
Abstract
Breast cancer will overtake all other cancers in terms of diagnoses in 2024. Breast cancer counts highest among women in terms of cancer incidence and death rates. Innovative treatment approaches are desperately needed because treatment resistance brought on by current clinical drugs impedes therapeutic efficacy. The T cell-based immunotherapy known as chimeric antigen receptor (CAR) T cell treatment, which uses the patient's immune cells to fight cancer, has demonstrated remarkable efficacy in treating hematologic malignancies; nevertheless, the treatment effects in solid tumors, like breast cancer, have not lived up to expectations. We discuss in detail the role of tumor-associated antigens in breast cancer, current clinical trials, barriers to the intended therapeutic effects of CAR-T cell therapy, and potential ways to increase treatment efficacy. Finally, our review aims to stimulate readers' curiosity by summarizing the most recent advancements in CAR-T cell therapy for breast cancer.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| | - Khaled Bedair
- Department of Social Sciences, College of Arts and Sciences, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| | - Layla Al-Mansoori
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Sara Caratelli
- Institute of Translational Pharmacology-CNR, Rome, Italy
| | | | - Alice Gaiba
- Institute of Translational Pharmacology-CNR, Rome, Italy
| | | |
Collapse
|
6
|
Gilden JK, Stecha P, Hartnett J, Cong M. A bioluminescent reporter bioassay for in-process assessment of chimeric antigen receptor lentiviral vector potency. Antib Ther 2025; 8:40-46. [PMID: 39839908 PMCID: PMC11744306 DOI: 10.1093/abt/tbae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/19/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025] Open
Abstract
Background Chimeric antigen receptor (CAR)-T-cell therapy is a breakthrough in the field of cancer immunotherapy, wherein T cells are genetically modified to recognize and attack cancer cells. Delivery of the CAR gene is a critical step in this therapy and is usually achieved by transducing patient T cells with a lentiviral vector (LV). Because the LV is an essential component of CAR-T manufacturing, there is a need for simple bioassays that reflect the mechanism of action (MOA) of the LV and can measure LV potency with accuracy and specificity. Common methods for LV quantification may overestimate functional titer and lack a functional readout of LV MOA. Methods We developed a bioluminescent reporter bioassay using Jurkat T cells stably expressing a luciferase reporter under the control of an nuclear factor of activated T cells (NFAT) response element and tested its suitability for measuring LV potency. Results Jurkat reporter cells can be transduced with CAR LV and combined with target cells, yielding a luminescent signal that is dependent on the identity and potency of the LV used. Bioluminescence was highly correlated with CAR expression. The assay is stability indicating and suitable for use in drug development and quality control settings. Conclusions We have developed a simple bioassay for potency testing of CAR LV. The bioassay represents a significant improvement over other approaches to LV quantification because it reflects the MOA of the LV and selectively detects fully functional viral particles, making it ideal for inclusion in a matrix of in-process quality control assays for CAR LV.
Collapse
Affiliation(s)
- Julia K Gilden
- Research and Development, Promega Corp., Madison, WI 53711, United States
| | - Pete Stecha
- Research and Development, Promega Corp., Madison, WI 53711, United States
| | - Jim Hartnett
- Research and Development, Promega Corp., Madison, WI 53711, United States
| | - Mei Cong
- Research and Development, Promega Corp., Madison, WI 53711, United States
| |
Collapse
|
7
|
Zeng L, Li Y, Xiang W, Xiao W, Long Z, Sun L. Advances in chimeric antigen receptor T cell therapy for autoimmune and autoinflammatory diseases and their complications. J Autoimmun 2025; 150:103350. [PMID: 39700677 DOI: 10.1016/j.jaut.2024.103350] [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: 05/29/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Chimeric antigen receptor T (CAR-T) cells are genetically engineered T cells expressing transmembrane chimeric antigen receptors with specific targeting abilities. As an emerging immunotherapy, the use of CAR-T cells has made significant breakthroughs in cancer treatment, particularly for hematological malignancies. The success of CAR-T cell therapy in blood cancers highlights its potential for other conditions in which the clearance of pathological cells is therapeutic, such as liver diseases, infectious diseases, heart failure, and diabetes. Given the limitations of current therapies for autoimmune diseases, researchers have actively explored the potential therapeutic value of CAR-T cells and their derivatives in the field of autoimmune diseases. This review focuses on the research progress and current challenges of CAR-T cells in autoimmune diseases with the aim of providing a theoretical basis for the precise treatment of autoimmune diseases. In the future, CAR-T cells may present new therapeutic modalities and ultimately provide hope for patients with autoimmune diseases.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Autoimmune Diseases/therapy
- Autoimmune Diseases/immunology
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- T-Lymphocytes/metabolism
- Animals
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Yan Li
- People's Hospital of Ningxiang City, Ningxiang City, China
| | - Wang Xiang
- Department of Rheumatology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changde City, China
| | - Wei Xiao
- Department of Rheumatology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changde City, China.
| | - Zhiyong Long
- Department of Physical Medicine and Rehabilitation, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China; Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
8
|
Diorio C, Teachey DT, Grupp SA. Allogeneic chimeric antigen receptor cell therapies for cancer: progress made and remaining roadblocks. Nat Rev Clin Oncol 2025; 22:10-27. [PMID: 39548270 DOI: 10.1038/s41571-024-00959-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 11/17/2024]
Abstract
Chimeric antigen receptor (CAR) T cells are revolutionizing cancer therapy, particularly for haematological malignancies, conferring durable and sometimes curative responses in patients with advanced-stage disease. The CAR T cell products currently approved for clinical use are all autologous and are often effective; however, in patients who are lymphopenic and/or heavily pretreated with chemotherapy, autologous T cells can be difficult to harvest in sufficient numbers or have functional impairments that might ultimately render them less efficacious. Moreover, autologous products take several weeks to produce, and each product can be used in only one patient. By contrast, allogeneic CAR T cells can be produced for many patients using T cells from a single healthy donor, can be optimized for safety and efficacy, can be instantly available for 'off-the-shelf' use and, therefore, might also be more cost-effective. Despite these potential advantages, the development of allogeneic CAR T cells has lagged behind that of autologous products, owing to the additional challenges such as avoiding graft-versus-host disease and host-mediated graft rejection. Over the past few years, the development of advanced genome-editing techniques has facilitated the generation of novel allogeneic CAR T cell products. Furthermore, CAR cell products derived from other cell types such as induced pluripotent stem cells and natural killer cells are being investigated for clinical use. In this Review, we discuss the potential of allogeneic CAR cell products to expand life-saving immunotherapy to a much broader population of patients in the coming years, the progress made to date and strategies to overcome remaining hurdles.
Collapse
Affiliation(s)
- Caroline Diorio
- Division of Oncology and Center for Childhood Cancer Research, Department of Paediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Susan S. and Stephen P. Kelly Center for Cancer Immunotherapy, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David T Teachey
- Division of Oncology and Center for Childhood Cancer Research, Department of Paediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Susan S. and Stephen P. Kelly Center for Cancer Immunotherapy, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stephan A Grupp
- Division of Oncology and Center for Childhood Cancer Research, Department of Paediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Susan S. and Stephen P. Kelly Center for Cancer Immunotherapy, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Wu Z, Wang Y, Jin X, Wang L. Universal CAR cell therapy: Challenges and expanding applications. Transl Oncol 2025; 51:102147. [PMID: 39413693 PMCID: PMC11525228 DOI: 10.1016/j.tranon.2024.102147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/20/2024] [Accepted: 09/28/2024] [Indexed: 10/18/2024] Open
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy has gained success in adoptive cell therapy for hematological malignancies. Although most CAR cell therapies in clinical trials or markets remain autologous, their acceptance has been limited due to issues like lengthy manufacturing, poor cell quality, and demanding cost. Consequently, "Off-the-shelf", universal CAR (UCAR) cell therapy has emerged. Current concerns with UCAR therapies revolve around side effects such as graft versus host disease (GVHD) and host versus graft response (HVGR). Preclinical research on UCAR cell therapies aims to enhance efficacy and minimize these side effects. Common approaches involve gene editing techniques to knock out T cell receptor (TCR), human leukocyte antigen (HLA), and CD52 expression to mitigate GVHD and HVGR risks. However, these methods carry drawbacks including potential genotoxicity of the edited cells. Most recently, novel editing techniques, such as epigenetic editing and RNA writer systems, have been developed to reduce the risk of GVHD and HVGR, allowing for multiplex editing at different sites. Additionally, incorporating more cell types into UCAR cell therapies, like T-cell subtypes (DNT, γδT, virus-specific T cells) and NK cells, can efficiently target tumors without triggering side effects. In addition, the limited efficacy of T cells and NK cells against solid tumors is being addressed through CAR-Macrophages. In summary, CAR cell therapy has evolved to accommodate multiple cell types while expanding applications to various diseases, including hematologic malignancies and solid tumors, which holds tremendous growth potential and is promised to improve the lives of more patients in the future.
Collapse
Affiliation(s)
- Ziyu Wu
- Department of Hematology I, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yifan Wang
- Department of Translational Medicine, Research Ward, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, China.
| | - Xin Jin
- Department of Translational Medicine, Research Ward, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, China.
| | - Luqiao Wang
- Department of Hematology I, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
10
|
Stein MN, Dumbrava EE, Teply BA, Gergis US, Guiterrez ME, Reshef R, Subudhi SK, Jacquemont CF, Senesac JH, Bayle JH, Scripture CD, Chatwal MS, Bilen MA, Stadler WM, Becerra CR. PSCA-targeted BPX-601 CAR T cells with pharmacological activation by rimiducid in metastatic pancreatic and prostate cancer: a phase 1 dose escalation trial. Nat Commun 2024; 15:10743. [PMID: 39737899 PMCID: PMC11685978 DOI: 10.1038/s41467-024-53220-6] [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: 07/18/2024] [Accepted: 10/07/2024] [Indexed: 01/01/2025] Open
Abstract
Here we report results of a phase 1 multi-institutional, open-label, dose-escalation trial (NCT02744287) of BPX-601, an investigational autologous PSCA-directed GoCAR-T® cell product containing an inducible MyD88/CD40 ON-switch responsive to the activating dimerizer rimiducid, in patients with metastatic pancreatic (mPDAC) or castration-resistant prostate cancer (mCRPC). Primary objectives were to evaluate safety and tolerability and determine the recommended phase 2 dose/schedule (RP2D). Secondary objectives included the assessment of efficacy and characterization of the pharmacokinetics of rimiducid. Thirty-three patients received BPX-601 with or without rimiducid, 24 patients with mPDAC and 9 with mCRPC. Two dose-limiting toxicities and two treatment-related deaths occurred in the highest-dose mCRPC cohort, after which the study was terminated, without determination of the RP2D. Two mCRPC patients experienced partial responses (one unconfirmed), and 56% of mCRPC patients achieved ≥50% reduction in prostate-specific antigen. BPX-601 cell expansion, long-term persistence in peripheral blood, and tumor infiltration were observed. Rimiducid increased circulating inflammatory cytokines/chemokines consistent with GoCAR-T® cell activation. These results suggest that pharmacological activation of GoCAR-T® cells is feasible and may offer a promising avenue to control chimeric antigen receptor-T cell activity with continued dose-optimization to improve tolerability.
Collapse
Affiliation(s)
- Mark N Stein
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | | | | | | | | | - Ran Reshef
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Sumit K Subudhi
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | - Monica S Chatwal
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Mehmet A Bilen
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | - Carlos R Becerra
- Baylor University Medical Center, Dallas, TX, USA
- Hoag Family Cancer Institute, Newport Beach, CA, USA
| |
Collapse
|
11
|
Ayoub M, Susin SA, Bauvois B. Tumor Cell Survival Factors and Angiogenesis in Chronic Lymphocytic Leukemia: How Hot Is the Link? Cancers (Basel) 2024; 17:72. [PMID: 39796700 PMCID: PMC11719013 DOI: 10.3390/cancers17010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of neoplastic CD5+/CD19+ B lymphocytes in the blood. These cells migrate to and proliferate in the bone marrow and lymphoid tissues. Despite the development of new therapies for CLL, drug resistance and disease relapse still occur; novel treatment approaches are therefore still needed. Inhibition of the angiogenesis involved in the progression of CLL might be a relevant therapeutic strategy. The literature data indicate that vascular endothelial growth factor, angiopoietin-2, and matrix metalloproteinase-9 are pro-angiogenic factors in CLL. A number of other CLL factors might have pro-angiogenic activity: fibroblast growth factor-2, certain chemokines (such as CXCL-12 and CXCL-2), tumor necrosis factor-α, insulin-like growth factor-1, neutrophil gelatinase-associated lipocalin, and progranulin. All these molecules contribute to the survival, proliferation, and migration of CLL cells. Here, we review the literature on these factors' respective expression profiles and roles in CLL. We also summarize the main results of preclinical and clinical trials of novel agents targeting most of these molecules in a CLL setting. Through the eradication of leukemic cells and the inhibition of angiogenesis, these therapeutic approaches might alter the course of CLL.
Collapse
Affiliation(s)
| | | | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS 1138, Drug Resistance in Hematological Malignancies Team, F-75006 Paris, France; (M.A.); (S.A.S.)
| |
Collapse
|
12
|
Di Napoli R, Balzano N, Ruggiero R, Mascolo A, Scavone C, di Mauro G, Capuano A. T-cell malignancies following CAR T-Cell therapy: insights from the FDA Adverse Event Reporting System (FAERS). Expert Opin Drug Saf 2024:1-10. [PMID: 39696955 DOI: 10.1080/14740338.2024.2443965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/10/2024] [Accepted: 10/18/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Concern about post-CAR T-cell lymphomas has recently emerged. Analysis of pharmacovigilance data contributes to continuous safety monitoring, especially for newly authorized medicines, like CAR-T therapies. RESEARCH DESIGN AND METHODS Individual case safety reports (ICSRs) reporting at least one CAR T-cell therapy as a suspect drug were extracted from the Food and Drug Administration Adverse Event Reporting System database up to 6 February 2024. Descriptive and disproportionality analysis were performed. RESULTS Seventeen ICSRs reported T-cell malignancies associated with CAR T-cell therapy. Gender distribution was similar between females and males, and adult patients accounted for 41.2% of ICSRs. All cases were serious, with 41.2% resulting in death. The most reported Preferred Terms (PTs) for T-cell malignancies was 'T-cell lymphoma' (70.6%). Over 70% of ICSRs reported at least one other adverse event, predominantly gastrointestinal disorders (14.3%). Axicabtagene ciloleucel and tisagenlecleucel were associated with a statistically higher reporting frequency of T-cell lymphoma compared to all other drugs (p-value <0.001, for both). Statistically higher reporting frequencies of 'Haematological malignant tumors' and 'Malignant lymphomas' SMQs emerged when tisagenlecleucel was compared with axicabtagene ciloleucel (p-value <0.001, for both). CONCLUSIONS Axicabtagene ciloleucel and tisagenlecleucel may be associated with a higher reporting frequency of T-cell lymphoma than other drugs.
Collapse
Affiliation(s)
- Raffaella Di Napoli
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
| | - Nunzia Balzano
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
| | - Rosanna Ruggiero
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Roma, Italy
| | - Annamaria Mascolo
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Roma, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Roma, Italy
| | - Gabriella di Mauro
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- UOC Pharmacy, AORN Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
| |
Collapse
|
13
|
Guo Q, Li J, Wang J, Li L, Wei J, Zhang L. The advent of chimeric antigen receptor T Cell therapy in recalibrating immune balance for rheumatic autoimmune disease treatment. Front Pharmacol 2024; 15:1502298. [PMID: 39734406 PMCID: PMC11672202 DOI: 10.3389/fphar.2024.1502298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
CAR-T cell therapy, a cutting-edge cellular immunotherapy with demonstrated efficacy in treating hematologic malignancies, also exhibits significant promise for addressing autoimmune diseases. This innovative therapeutic approach holds promise for achieving long-term remission in autoimmune diseases, potentially offering significant benefits to affected patients. Current targets under investigation for the treatment of these conditions include CD19, CD20, and BCMA, among others. However, CAR-T therapy faces difficulties such as time-consuming cell manufacturing, complex and expensive process, and the possibility of severe adverse reactions complicating the treatment, etc. This article examines CAR-T therapy across various rheumatic autoimmune diseases, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Sjögren's syndrome (SS), systemic sclerosis (SSc), antisynthetase syndrome (ASS), and ANCA-associated vasculitis (AAV), highlighting both therapeutic advancements and ongoing challenges.
Collapse
Affiliation(s)
- Qianyu Guo
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Jie Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Juanjuan Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Linxin Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jia Wei
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
14
|
Huang R, Chen H, Xie J, Lou Q, Tan L, Zhang N, An Z, John S, Zhang CC. A Switch Protein Adapter for Anti-LILRB4 CAR-T Cells. Eur J Immunol 2024:eji5889. [PMID: 39663681 DOI: 10.1002/eji.202451172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 11/14/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024]
Abstract
Chimeric antigen receptor-T cell (CAR-T) immunotherapy has shown remarkable results for the treatment of certain hematologic malignancies. A redirection strategy that utilizes clinically relevant CAR-T cells in combination with adapter proteins may be an effective strategy to target other hematologic and solid cancers. We established a fusion antibody-based strategy with flexibility to target multiple tumor types in combination with a novel anti-leukocyte immunoglobulin-like receptor-B 4 (LILRB4) CAR-T cell. Specifically, we engineered switch protein (SwP) adapters containing the LILRB4 extracellular domain fused to either an anti-CD19 or anti-CD20 single-chain variable fragment (scFv). These SwPs were sufficient to stimulate anti-LILRB4 CAR-T cells against SwP-tagged LILRB4-CD19+ and LILRB4-CD20+ cancers in vitro and in vivo. This strategy may allow CAR-T cells to be redirected against a variety of tumor antigens and cancer types and become a valuable approach to expand the impact of cellular immunotherapy.
Collapse
Affiliation(s)
- Ryan Huang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Heyu Chen
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jingjing Xie
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Qi Lou
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lingxiao Tan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Samuel John
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Cheng Cheng Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
15
|
Sainatham C, Yadav D, Dilli Babu A, Tallapalli JR, Kanagala SG, Filippov E, Murillo Chavez F, Ahmed N, Lutfi F. The current socioeconomic and regulatory landscape of immune effector cell therapies. Front Med (Lausanne) 2024; 11:1462307. [PMID: 39697210 PMCID: PMC11652178 DOI: 10.3389/fmed.2024.1462307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Immune cell effector therapies, including chimeric antigen receptor (CAR)-T cells, T-cell receptor (TCR) T cells, natural killer (NK) cells, and macrophage-based therapies, represent a transformative approach to cancer treatment, harnessing the immune system to target and eradicate malignant cells. CAR-T cell therapy, the most established among these, involves engineering T cells to express CARs specific to cancer cell antigens, showing remarkable efficacy in hematologic malignancies like leukemias, B-cell lymphomas, and multiple myeloma. Similarly, TCR-modified therapies, which reprogram T cells to recognize intracellular tumor antigens presented by major histocompatibility complex (MHC) molecules, offer promise for a range of solid tumors. NK-cell therapies leverage NK cells' innate cytotoxicity, providing an allogeneic approach that avoids some of the immune-related complications associated with T-cell-based therapies. Macrophage-based therapies, still in early stages of the development, focus on reprogramming macrophages to stimulate an immune response against cancer cells in the tumor microenvironment. Despite their promise, socioeconomic and regulatory challenges hinder the accessibility and scalability of immune cell effector therapies. These treatments are costly, with CAR-T therapies currently exceeding $400,000 per patient, creating significant disparities in access based on socioeconomic status and geographic location. The high manufacturing costs stem from the personalized, labor-intensive processes of harvesting, modifying, and expanding patients' cells. Moreover, complex logistics for manufacturing and delivering these therapies limit their reach, particularly in low-resource settings. Regulatory pathways further complicate the landscape. In the United States., the Food and Drug Administrations' (FDA) accelerated approval processes for cell-based therapies facilitate innovation but do not address cost-related barriers. In Europe, the European Medicines Agency (EMA) offers adaptive pathways, yet decentralized reimbursement systems create uneven access across member states. Additionally, differing regulatory standards for manufacturing and quality control worldwide pose hurdles for global harmonization and access. To expand the reach of immune effector cell therapies, a multipronged approach is needed-streamlined regulatory frameworks, policies to reduce treatment costs, and international collaborations to standardize manufacturing. Addressing these socioeconomic and regulatory obstacles is essential to make these life-saving therapies accessible to a broader patient population worldwide. We present a literature review on the current landscape of immune effector cell therapies and barriers of access to currently approved standard of care therapy at various levels.
Collapse
Affiliation(s)
- Chiranjeevi Sainatham
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Devvrat Yadav
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Aravind Dilli Babu
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Jayanth Reddy Tallapalli
- Division of Infectious Diseases, Department of Internal Medicine, University of South Florida, Tampa, FL, United States
| | - Sai Gautham Kanagala
- Department of Internal Medicine, New York Medical College/Metropolitan Hospital Center, New York, NY, United States
| | - Evgenii Filippov
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Franco Murillo Chavez
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Nausheen Ahmed
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Forat Lutfi
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
16
|
Huntington SF, Lin D, Lafeuille M, Thompson‐Leduc P, Shah A, Kim N, Hester L, Tardif‐Samson A, Moore B, Fowler J, Marshall A, Zhang X, Gifkins D, Wu B. Identification of cytokine release syndrome and indicators of severity in retrospective databases among patients receiving immunotherapy. Pharmacol Res Perspect 2024; 12:e70024. [PMID: 39487576 PMCID: PMC11530407 DOI: 10.1002/prp2.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/05/2024] [Accepted: 09/24/2024] [Indexed: 11/04/2024] Open
Abstract
Cytokine release syndrome (CRS) can occur following cancer immunotherapies, but is most often mild and of limited duration. International Classification of Diseases (ICD)-10 codes allowing identification of CRS were introduced in 2020 but may be underutilized. We evaluated the performance of a published claims-based algorithm to detect CRS (any grade) and high-grade CRS (HG, grades 2-5), as well as identified indicators of HG CRS in retrospective data. Adults with low-grade and HG CRS during an encounter coinciding with administrations of blinatumomab or chimeric antigen receptor-T therapy were identified in three types of retrospective databases (hospital chargemaster data, electronic health records, and administrative claims). The algorithm's sensitivity in detecting any CRS and HG CRS was reported. A least absolute shrinkage and selection operator (LASSO) regression model was developed to identify indicators of HG CRS. Performance of the model was evaluated using area under the curve (AUC). The sensitivity of the algorithm to detect any grade CRS ranged between 77%-100% and between 8%-80% for HG CRS, depending on the type of database. The LASSO model identified hypotension, positive pressure (including mechanical ventilation), tocilizumab, and vasopressors as indicators of HG CRS. AUC varied between 60% and 75%. The algorithm accurately detected any grade CRS for over three-quarters of instances, but was not as reliable for HG CRS. Results varied based on database attributes. Hypotension, vasopressors, positive pressure, and tocilizumab were associated with HG CRS and may be methodologically helpful signals of CRS severity in retrospective data.
Collapse
Affiliation(s)
| | - Dee Lin
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| | | | | | - Aditi Shah
- Analysis Group, Inc.MontrealQuebecCanada
| | - Nina Kim
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| | - Laura Hester
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| | | | | | - Jessica Fowler
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| | - Alexander Marshall
- Janssen Global Services, LLC, a Johnson and Johnson CompanyRaritanPennsylvaniaUSA
| | - Xinke Zhang
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| | - Dina Gifkins
- Janssen Research and Development, LLC, a Johnson and Johnson CompanyRaritanPennsylvaniaUSA
| | - Bingcao Wu
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| |
Collapse
|
17
|
Rocka A, Suchcicka M, Jankowska AM, Woźniak MM, Lejman M. Pathway of LCK Tyrosine Kinase and mTOR Signaling in Children with T-Cell Acute Lymphoblastic Leukemia. Appl Clin Genet 2024; 17:187-198. [PMID: 39583285 PMCID: PMC11585986 DOI: 10.2147/tacg.s494389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/04/2024] [Indexed: 11/26/2024] Open
Abstract
The aim of this study is to analyze available research on targeting signaling pathways for the development of new drugs in patients with T-cell acute lymphoblastic leukemia (T-ALL). This analysis focuses specifically on the role of LCK tyrosine kinase and mTOR signaling pathways in pediatric patients. Outcome: Current literature suggests that these pathways play a significant role in the regulation of T-cell cycles, making them potential therapeutic targets. However, despite promising findings, there remains a need for further research, particularly in pediatric populations, to fully understand the therapeutic implications and to optimize drug development. The conclusion drawn from this analysis highlights the significant influence of LCK and mTOR on T-cell cycle regulation, underscoring the importance of continued investigation in this area.
Collapse
Affiliation(s)
- Agata Rocka
- Pediatric Radiology, Medical University of Lublin, Medical University of Lublin, Prof. Antoni Gębali 6, Lublin, 20-093, Poland
| | - Maria Suchcicka
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, Prof. Antoni Gębali 6, Lublin, 20-093, Poland
| | - Aleksandra M Jankowska
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, Prof. Antoni Gębali 6, Lublin, 20-093, Poland
| | - Magdalena M Woźniak
- Pediatric Radiology, Medical University of Lublin, Medical University of Lublin, Prof. Antoni Gębali 6, Lublin, 20-093, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, Prof. Antoni Gębali 6, Lublin, 20-093, Poland
| |
Collapse
|
18
|
Tigu AB, Munteanu R, Moldovan C, Rares D, Kegyes D, Tomai R, Moisoiu V, Ghiaur G, Tomuleasa C, Einsele H, Gulei D, Croce CM. Therapeutic advances in the targeting of ROR1 in hematological cancers. Cell Death Discov 2024; 10:471. [PMID: 39551787 PMCID: PMC11570672 DOI: 10.1038/s41420-024-02239-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024] Open
Abstract
Receptor tyrosine kinases (RTKs) are key cell surface receptors involved in cell communication and signal transduction, with great importance in cell growth, differentiation, survival, and metabolism. Dysregulation of RTKs, such as EGFR, VEGFR, HER2 or ROR, could lead to various diseases, particularly cancers. ROR1 has emerged as a promising target in hematological malignancies. The development of ROR1 targeted therapies is continuously growing leading to remarkable novel therapeutical approaches using mAbs, antibody-drug conjugates, several small molecules or CAR T cells which have shown encouraging preclinical results. In the hematological field, mAbs, small molecules, BiTEs or CAR T cell therapies displayed promising outcomes with the clinical trials data encouraging the use of anti-ROR1 therapies. This paper aims to offer a comprehensive analysis of the current landscape of ROR1-targeted therapies in hematological malignancies marking the innovative approaches with promising preclinical and clinical. Offering a better understanding of structural and functional aspects of ROR1 could lead to new perspectives in targeting a wide spectrum of malignancies.
Collapse
Affiliation(s)
- Adrian-Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy Cluj Napoca, Cluj Napoca, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy Cluj Napoca, Cluj Napoca, Romania
| | - Cristian Moldovan
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy Cluj Napoca, Cluj Napoca, Romania
| | - Drula Rares
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Kegyes
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy Cluj Napoca, Cluj Napoca, Romania
| | - Radu Tomai
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy Cluj Napoca, Cluj Napoca, Romania
| | - Vlad Moisoiu
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Gabriel Ghiaur
- Division of Hematological Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy Cluj Napoca, Cluj Napoca, Romania.
| | - Hermann Einsele
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy Cluj Napoca, Cluj Napoca, Romania
- Department of Medicine, University of Würzburg, Würzburg, Germany
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy Cluj Napoca, Cluj Napoca, Romania
| | - Carlo M Croce
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
19
|
Massariol Pimenta T, Carlos de Souza J, da Silva Martins B, Silva Butzene SM, Simões Padilha JM, Ganho Marçal M, Dos Santos Elias G, Rangel LBA. Emerging strategies to overcome ovarian cancer: advances in immunotherapy. Front Pharmacol 2024; 15:1490896. [PMID: 39564107 PMCID: PMC11573523 DOI: 10.3389/fphar.2024.1490896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
Ovarian cancer is the second most common malignant neoplasm of gynecological origin and the leading cause of death from cancer in the female reproductive system worldwide. This scenario is largely due to late diagnoses, often in advanced stages, and the development of chemoresistance by cancer cells. These challenges highlight the need for alternative treatments, with immunotherapy being a promising option. Cancer immunotherapy involves triggering an anti-tumor immune response and developing immunological memory to eliminate malignant cells, prevent recurrence, and inhibit metastasis. Some ongoing research investigate potentially immunological advancements in the field of cancer vaccines, immune checkpoint blockade, CAR-T cell, and other strategies.
Collapse
Affiliation(s)
- Tatiana Massariol Pimenta
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Josiany Carlos de Souza
- Biotechnology Program/RENORBIO, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Bárbara da Silva Martins
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Solenny Maria Silva Butzene
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - José Matheus Simões Padilha
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Milleny Ganho Marçal
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Guilherme Dos Santos Elias
- Biochemistry Program, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Leticia Batista Azevedo Rangel
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
- Biotechnology Program/RENORBIO, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
- Biochemistry Program, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
20
|
Lungova K, Putman M. Barriers to CAR T-cell therapy in rheumatology. THE LANCET. RHEUMATOLOGY 2024:S2665-9913(24)00240-6. [PMID: 39515366 DOI: 10.1016/s2665-9913(24)00240-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 11/16/2024]
Abstract
Chimeric antigen receptor (CAR) T cells have recently shown remarkable promise in treating rheumatic diseases, including systemic lupus erythematosus (SLE), idiopathic inflammatory myopathies, and systemic sclerosis. Currently, there are 37 clinical trials registered for CAR T-cell therapy in rheumatic diseases and many more are being planned. Much of this enthusiasm is justifiable, but widespread adoption of CAR T-cell therapy in rheumatology faces several barriers. The trajectory of autoimmune diseases differs from malignancies and a surprisingly narrow population could be eligible for CAR T-cell therapy. Current CAR T-cell approaches rely on B-cell depletion, which has a mixed record of success for many diseases. The high cost of CAR T-cell therapy and potential safety concerns, such as cytokine release syndrome and long-term infection risks, also pose substantial challenges. Moving forward, more targeted CAR T-cell approaches, such as antigen-specific chimeric autoantibody receptors or chimeric autoantigen T-cell receptors, could offer greater efficacy and safety in treating rheumatic diseases.
Collapse
Affiliation(s)
- Karolina Lungova
- Division of Rheumatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael Putman
- Division of Rheumatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
21
|
Sutherland NM, Zhou B, Zhang L, Ong MS, Hong JS, Pak A, Liu KJ, Frigault MJ, Maus MV, Hill JA, Reynolds K, Walter JE, Camargo CA, Barmettler S. Association of CD19 +-targeted chimeric antigen receptor (CAR) T-cell therapy with hypogammaglobulinemia, infection, and mortality. J Allergy Clin Immunol 2024:S0091-6749(24)01165-5. [PMID: 39505278 DOI: 10.1016/j.jaci.2024.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND CD19-targeted chimeric antigen receptor T-cell therapy (CAR-T therapy) has revolutionized the treatment of hematologic malignancies. As these cells target CD19+ receptors on B cells, there is the potential for B-cell aplasia and hypogammaglobulinemia. Data on the degree and clinical significance of hypogammaglobulinemia are sparse. OBJECTIVES We sought to evaluate hypogammaglobulinemia after CD19-targeted CAR-T therapy and risk factors for hypogammaglobulinemia, infections, and mortality. METHODS We performed a retrospective evaluation of 579 patients receiving CD19-directed CAR-T therapy and evaluated demographics, hypogammaglobulinemia (IgG ≤600 mg/dL), infections prior to and after CAR-T therapy, and risk factors for hypogammaglobulinemia, infection, hospitalizations, and mortality. RESULTS Patients had a mean age of 64 years and 64% were male. Prior to CAR-T therapy, 60% of patients had hypogammaglobulinemia, which increased to 91% post-CAR-T therapy. Mean IgG levels decreased from pre- to post-CAR-T therapy levels (587 to 362 mg/dL; P < .0001). Thirty-seven percent of patients developed a serious infection post-CAR-T therapy. Hypogammaglobulinemia prior to CAR-T therapy was associated with worsening hypogammaglobulinemia after CAR-T therapy. Hypogammaglobulinemia post CAR-T therapy was associated with an increased risk of serious infection following CAR-T therapy (incidence rate ratio: 2.7; 95% CI: 1.5-5.2; P = .002). Risk factors for mortality included mild hypogammaglobulinemia (400 mg/dL < IgG ≤ 600 mg/dL), infections ≤100 days post-CAR-T therapy, and hospitalizations for infections. Immunoglobulin replacement was associated with a decreased risk of mortality. CONCLUSIONS We identified ∼90% of patients with hypogammaglobulinemia after CAR-T therapy. Hypogammaglobulinemia before CAR-T therapy was strongly predictive of worsening hypogammaglobulinemia after CAR-T therapy, which was associated with an increased risk of serious infection and mortality post CAR-T therapy. Increased immunological monitoring is needed to identify high-risk patients who may benefit from interventions to decrease morbidity and mortality.
Collapse
Affiliation(s)
| | - Baijun Zhou
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Lingxiao Zhang
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Mei-Sing Ong
- Department of Population Medicine, Harvard Medical School, Boston, Mass; Harvard Pilgrim Health Care Institute, Boston, Mass
| | - Joseph S Hong
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Andrew Pak
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Katherine J Liu
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Matthew J Frigault
- Harvard Medical School, Boston, Mass; Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Marcela V Maus
- Harvard Medical School, Boston, Mass; Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, Mass
| | - Joshua A Hill
- Department of Medicine, University of Washington, Seattle, Wash; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Wash; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Wash; Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, Wash
| | - Kerry Reynolds
- Harvard Medical School, Boston, Mass; Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Jolan E Walter
- Department of Pediatrics, University of South Florida, St Petersburg, Fla; Department of Medicine, University of South Florida, St Petersburg, Fla; Division of Pediatric Allergy/Immunology, Johns Hopkins-All Children's Hospital, St Petersburg, Fla; Division of Pediatric Allergy Immunology, Massachusetts General Hospital, Boston, Mass
| | - Carlos A Camargo
- Harvard Medical School, Boston, Mass; Department of Emergency Medicine, Massachusetts General Hospital, Boston, Mass
| | - Sara Barmettler
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass.
| |
Collapse
|
22
|
Rabie LE, Mohran AA, Gaber KA, Ali NM, Abd El Naby AM, Ghoniem EA, Abd Elmaksod BA, Abdallah AN. Beyond Conventional Treatments: Exploring CAR-T Cell Therapy for Cancer Stem Cell Eradication. Stem Cell Rev Rep 2024; 20:2001-2015. [PMID: 39312080 PMCID: PMC11554798 DOI: 10.1007/s12015-024-10786-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 11/12/2024]
Abstract
BACKGROUND For decades cancer remained the center of attention in the scientific community as its survival rates are low. Researchers from all around the world wanted to know the core of the problem as to what initiates cancer in a patient and helps with its progression. Many postulations came to light, but Cancer Stem Cells (CSC) was the most appealing and convincing. MAIN BODY In this review, we shed light on a potential solution to the problem by reviewing CAR-T cells (Chimeric antigen receptor T cells). These specialized T cells are designed to detect specific antigens on cancer cells. We analyse the steps of their formation from the collection of T cells from the patient's bloodstream and modifying it to exhibit specific CAR structures on their surfaces, to reinjecting them back and evaluating their efficacy. We thoroughly investigate the structure of the CAR design with improvements across different generations. The focus extends to the unique properties of CSCs as in how targeting specific markers on them can enhance the precision of cancer therapy. CONCLUSION Despite the successes, the review discusses the existing limitations and toxicities associated with CAR-derived therapies, highlighting the ongoing need for research and refinement. Looking ahead, we explore proposed strategies aimed at optimizing CAR-T cell therapy to mitigate adverse effects for improved cancer treatments.
Collapse
Affiliation(s)
- Lobna E Rabie
- Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Ahmed A Mohran
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Kholoud A Gaber
- Molecular Biology and Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Nour M Ali
- Chemistry Department, Faculty of Science, KFS University, Kafr El-Sheikh, Egypt
| | - Asmaa M Abd El Naby
- Zoology-Chemistry Department, Faculty of Science, Beni Suef University, Beni Suef, Egypt
| | - Eman A Ghoniem
- Biotechnology and Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | | | - Ahmed N Abdallah
- Hormones Department, Medical Research and Clinical Studies Institute, National research Centre, Cairo, Egypt
| |
Collapse
|
23
|
Malhotra J, Huang A, Amini A, Lee P. Novel Immunotherapeutics for the Treatment of Non-Small Cell Lung Cancer (NSCLC) Resistant to PD-1/PD-L1 Inhibitors. Cancers (Basel) 2024; 16:3603. [PMID: 39518043 PMCID: PMC11545025 DOI: 10.3390/cancers16213603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Immunotherapy with PD-1/PD-L1 inhibitors is the standard method of care for the treatment of newly diagnosed advanced or metastatic NSCLC, with or without chemotherapy. Many tumors, however, develop resistance to these immunotherapy agents. There is a need to develop more effective therapies for patients with metastatic NSCLC in the second-line setting and beyond. In this review, we present an overview of novel immunotherapies being investigated regarding the treatment of these patients. We summarize completed, as well as ongoing, trials investigating these therapies as monotherapy or in combination with PD-1/PD-L1 inhibitors. These include immune co-stimulatory antibodies, T-cell agonists, oncolytic viruses, vaccines, TIL therapies, and CAR-T therapies.
Collapse
Affiliation(s)
- Jyoti Malhotra
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Amy Huang
- Department of Medicine, University of Connecticut School of Medicine, Farmington, CT 06030, USA;
| | - Arya Amini
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.A.); (P.L.)
| | - Percy Lee
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.A.); (P.L.)
| |
Collapse
|
24
|
Chhabra R. Molecular and modular intricacies of precision oncology. Front Immunol 2024; 15:1476494. [PMID: 39507541 PMCID: PMC11537923 DOI: 10.3389/fimmu.2024.1476494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Precision medicine is revolutionizing the world in combating different disease modalities, including cancer. The concept of personalized treatments is not new, but modeling it into a reality has faced various limitations. The last decade has seen significant improvements in incorporating several novel tools, scientific innovations and governmental support in precision oncology. However, the socio-economic factors and risk-benefit analyses are important considerations. This mini review includes a summary of some commendable milestones, which are not just a series of successes, but also a cautious outlook to the challenges and practical implications of the advancing techno-medical era.
Collapse
Affiliation(s)
- Ravneet Chhabra
- Business Department, Biocytogen Boston Corporation, Waltham, MA, United States
| |
Collapse
|
25
|
Blount SL, Liu X, McBride JD. The Utilization of PRAME in the Diagnosis, Prognosis, and Treatment of Melanoma. Cells 2024; 13:1740. [PMID: 39451258 PMCID: PMC11505691 DOI: 10.3390/cells13201740] [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: 09/16/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Melanoma, a deadly form of skin cancer, has seen improved survival rates due to advances in diagnosis and treatment, yet the need for further improvement remains critical. Tumor-associated antigens, such as PRAME (Preferentially Expressed Antigen in Melanoma), offer promising avenues for enhanced diagnostic precision, prognostic assessment, and targeted immunotherapy. PRAME, a cancer testis antigen, is selectively expressed in various cancers, including melanoma, and plays a key role in promoting tumorigenesis through inhibition of retinoic acid signaling, epithelial-to-mesenchymal transition, and immune evasion. This review explores the diagnostic utility of PRAME in distinguishing melanoma from benign nevi, its prognostic value in aggressive melanoma subtypes, and its potential as a therapeutic target in cancer vaccines and adoptive T-cell therapies. While PRAME-targeted therapies face challenges such as tumor heterogeneity and immune suppression, ongoing research aims to overcome these barriers, offering hope for more effective melanoma treatments.
Collapse
Affiliation(s)
- Samuel L. Blount
- College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Xiaochen Liu
- Department of Dermatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Jeffrey D. McBride
- College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Dermatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| |
Collapse
|
26
|
Stilpeanu RI, Secara BS, Cretu-Stancu M, Bucur O. Oncolytic Viruses as Reliable Adjuvants in CAR-T Cell Therapy for Solid Tumors. Int J Mol Sci 2024; 25:11127. [PMID: 39456909 PMCID: PMC11508774 DOI: 10.3390/ijms252011127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Although impactful scientific advancements have recently been made in cancer therapy, there remains an opportunity for future improvements. Immunotherapy is perhaps one of the most cutting-edge categories of therapies demonstrating potential in the clinical setting. Genetically engineered T cells express chimeric antigen receptors (CARs), which can detect signals expressed by the molecules present on the surface of cancer cells, also called tumor-associated antigens (TAAs). Their effectiveness has been extensively demonstrated in hematological cancers; therefore, these results can establish the groundwork for their applications on a wide range of requirements. However, the application of CAR-T cell technology for solid tumors has several challenges, such as the existence of an immune-suppressing tumor microenvironment and/or inadequate tumor infiltration. Consequently, combining therapies such as CAR-T cell technology with other approaches has been proposed. The effectiveness of combining CAR-T cell with oncolytic virus therapy, with either genetically altered or naturally occurring viruses, to target tumor cells is currently under investigation, with several clinical trials being conducted. This narrative review summarizes the current advancements, opportunities, benefits, and limitations in using each therapy alone and their combination. The use of oncolytic viruses offers an opportunity to address the existing challenges of CAR-T cell therapy, which appear in the process of trying to overcome solid tumors, through the combination of their strengths. Additionally, utilizing oncolytic viruses allows researchers to modify the virus, thus enabling the targeted delivery of specific therapeutic agents within the tumor environment. This, in turn, can potentially enhance the cytotoxic effect and therapeutic potential of CAR-T cell technology on solid malignancies, with impactful results in the clinical setting.
Collapse
MESH Headings
- Humans
- Neoplasms/therapy
- Neoplasms/immunology
- Oncolytic Viruses/genetics
- Oncolytic Viruses/immunology
- Immunotherapy, Adoptive/methods
- Oncolytic Virotherapy/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Animals
- Tumor Microenvironment/immunology
- T-Lymphocytes/immunology
- Combined Modality Therapy/methods
- Adjuvants, Immunologic
- Antigens, Neoplasm/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Ruxandra Ilinca Stilpeanu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
| | - Bianca Stefania Secara
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
| | | | - Octavian Bucur
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
- Genomics Research and Development Institute, 020021 Bucharest, Romania
- Viron Molecular Medicine Institute, Boston, MA 02108, USA
| |
Collapse
|
27
|
Gavriilaki E, Demosthenous C, Evangelidis P, Bousiou Z, Batsis I, Vardi A, Mallouri D, Koravou EE, Spyridis N, Panteliadou A, Karavalakis G, Masmanidou M, Touloumenidou T, Papalexandri A, Poziopoulos C, Yannaki E, Sakellari I, Politou M, Papassotiriou I. Soluble Urokinase-Type Plasminogen Activator Receptor (suPAR), Growth Differentiation Factor-15 (GDF-15), and Soluble C5b-9 (sC5b-9) Levels Are Significantly Associated with Endothelial Injury Indices in CAR-T Cell Recipients. Int J Mol Sci 2024; 25:11028. [PMID: 39456810 PMCID: PMC11507105 DOI: 10.3390/ijms252011028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Endothelial injury indices, such as Endothelial Activation and Stress Index (EASIX), modified EASIX (m-EASIX), and simplified EASIX (s-EASIX) scores, have been previously associated with chimeric antigen receptor-T (CAR-T) cell immunotherapy complications. Soluble urokinase-type plasminogen activator receptor (suPAR), growth differentiation factor-15 (GDF-15), and soluble C5b-9 (sC5b-9) have been described as markers of endothelial injury post-hematopoietic stem cell transplantation. In the current study, we examined whether suPAR, GDF-15, and sC5b-9 levels were associated with endothelial injury indices in adult CAR-T cell recipients. The levels of these markers were measured in patients before CAR-T cell infusion and in healthy individuals with immunoenzymatic methods. We studied 45 CAR-T cell recipients and 20 healthy individuals as the control group. SuPAR, GDF-15, and sC5b-9 levels were significantly higher in the patients' group compared to the healthy control group (p < 0.001, in all comparisons). SuPAR levels at baseline were associated with the m-EASIX scores calculated at the same time point (p = 0.020), while suPAR and GDF-15 concentrations were correlated with EASIX scores at day 14 post-infusion (p < 0.001 in both comparisons). Moreover, sC5b-9 levels were correlated with the s-EASIX scores at infusion (p = 0.008) and the EASIX scores at day 14 (p = 0.005). In our study, sC5b9, suPAR, and GDF-15 levels were found to reflect endothelial injury in CAR-T cell recipients.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Christos Demosthenous
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Paschalis Evangelidis
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Zoi Bousiou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Ioannis Batsis
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Anna Vardi
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Despina Mallouri
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Eudoxia-Evaggelia Koravou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Nikolaos Spyridis
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Alkistis Panteliadou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Georgios Karavalakis
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Marianna Masmanidou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Tasoula Touloumenidou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Apostolia Papalexandri
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | | | - Evangelia Yannaki
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Ioanna Sakellari
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Marianna Politou
- Thrombosis–Bleeding–Transfusion Medicine Postgraduate Studies, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Hematology Laboratory-Blood Bank, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Papassotiriou
- First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
28
|
Looi CK, Loo EM, Lim HC, Chew YL, Chin KY, Cheah SC, Goh BH, Mai CW. Revolutionizing the treatment for nasopharyngeal cancer: the impact, challenges and strategies of stem cell and genetically engineered cell therapies. Front Immunol 2024; 15:1484535. [PMID: 39450176 PMCID: PMC11499120 DOI: 10.3389/fimmu.2024.1484535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a distinct malignancy of the nasopharynx and is consistently associated with the Epstein-Barr virus (EBV) infection. Its unique anatomical location and complex aetiology often result in advanced-stage disease at first diagnosis. While radiotherapy (RT) and chemotherapy have been the mainstays of treatment, they often fail to prevent tumour recurrence and metastasis, leading to high rates of treatment failure and mortality. Recent advancement in cell-based therapies, such as chimeric antigen receptor (CAR)-T cell therapy, have shown great promise in hematological malignancies and are now being investigated for NPC. However, challenges such as targeting specific tumour antigens, limited T cell persistence and proliferation, and managing treatment-related toxicities must be addressed. Extensive research is needed to enhance the effectiveness and safety of these therapies, paving the way for their integration into standard clinical practice for better management of NPC and a better quality of life for human health.
Collapse
Affiliation(s)
- Chin-King Looi
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Ee-Mun Loo
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
- Advanced Genomics Laboratory, AGTC Genomics, Kuala Lumpur, Malaysia
| | - Heng-Chee Lim
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Yik-Ling Chew
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shiau-Chuen Cheah
- Faculty of Medicine and Health Sciences, UCSI University, Port Dickson, Negeri Sembilan, Malaysia
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre, School of Medical and Life Sciences, Sunway University Malaysia, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, China
| | - Chun-Wai Mai
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
29
|
Elsemary MT, Maritz MF, Smith LE, Warkiani ME, Thierry B. Enrichment of T-lymphocytes from leukemic blood using inertial microfluidics toward improved chimeric antigen receptor-T cell manufacturing. Cytotherapy 2024; 26:1264-1274. [PMID: 38819362 DOI: 10.1016/j.jcyt.2024.05.005] [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: 02/29/2024] [Revised: 04/30/2024] [Accepted: 05/05/2024] [Indexed: 06/01/2024]
Abstract
Chimeric antigen receptor cell therapy is a successful immunotherapy for the treatment of blood cancers. However, hurdles in their manufacturing remain including efficient isolation and purification of the T-cell starting material. Herein, we describe a one-step separation based on inertial spiral microfluidics for efficient enrichment of T-cells in B-cell acute lymphoblastic leukemia (ALL) and B-cell chronic lymphocytic leukemia patient's samples. In healthy donors used to optimize the process, the lymphocyte purity was enriched from 65% (SD ± 0.2) to 91% (SD ± 0.06) and T-cell purity was enriched from 45% (SD ± 0.1) to 73% (SD ± 0.02). Leukemic samples had higher starting B-cells compared to the healthy donor samples. Efficient enrichment and recovery of lymphocytes and T-cells were achieved in ALL samples with B-cells, monocytes and leukemic blasts depleted by 80% (SD ± 0.09), 89% (SD ± 0.1) and 74% (SD ± 0.09), respectively, and a 70% (SD ± 0.1) T-cell recovery. Chronic lymphocytic leukemia samples had lower T-cell numbers, and the separation process was less efficient compared to the ALL. This study demonstrates the use of inertial microfluidics for T-cell enrichment and depletion of B-cell blasts in ALL, suggesting its potential to address a key bottleneck of the chimeric antigen receptor-T manufacturing workflow.
Collapse
MESH Headings
- Humans
- T-Lymphocytes/immunology
- Receptors, Chimeric Antigen/immunology
- Immunotherapy, Adoptive/methods
- Microfluidics/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Cell Separation/methods
- B-Lymphocytes/immunology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Mona T Elsemary
- Future Industries Institute, University of South Australia Mawson Lakes Campus, Mawson Lakes, SA, Australia
| | - Michelle F Maritz
- Future Industries Institute, University of South Australia Mawson Lakes Campus, Mawson Lakes, SA, Australia
| | - Louise E Smith
- Future Industries Institute, University of South Australia Mawson Lakes Campus, Mawson Lakes, SA, Australia
| | | | - Benjamin Thierry
- Future Industries Institute, University of South Australia Mawson Lakes Campus, Mawson Lakes, SA, Australia.
| |
Collapse
|
30
|
Khatsuria F, McMullan C, Aiyegbusi OL, Shaw KL, Iqbal R, Kinsella F, Wilson K, Pyatt L, Lewis M, Wheldon SMR, Burns D, Chakraverty R, Calvert M, Hughes SE. Development of a conceptual framework for an electronic patient-reported outcome (ePRO) system measuring symptoms and impacts of CAR T-cell therapies in patients with haematological malignancies. Lancet Oncol 2024; 25:e476-e488. [PMID: 39362259 DOI: 10.1016/s1470-2045(24)00256-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 10/05/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is associated with potentially severe toxicities that create a substantial burden for patients. Patient-reported outcomes (PROs) offer valuable insights into symptoms, functioning, and other complex constructs of interest. In this Review, we aimed to identify symptom and impact concepts important to patients receiving CAR T-cell therapy, construct a conceptual framework for an electronic patient-reported outcome (ePRO) system, and identify timepoints to capture PRO data for CAR T-cell therapies. We searched MEDLINE (OVID) and Web of Science (Clarivate) for articles in English published from Aug 30, 2017, to March 2, 2023. No restrictions on study design were applied. 178 symptoms or constructs were extracted from 44 articles reporting PRO collection in adults with haematological malignancies receiving CAR T-cell therapy. Six health-care professionals and 11 patients and caregiver partners verified construct relevance to clinical management and lived experience, respectively. 109 constructs were sorted according to the four domains of conceptual framework: symptom burden, impact of disease and treatment, tolerability, and health-related quality of life. The identification of concepts beyond symptom burden underscores the importance of PRO measurement for long-term monitoring, to align outcomes with patient concerns. The framework will facilitate PRO measure selection for systematic gathering of PROs from individuals with haematological malignancies receiving CAR T-cell therapies.
Collapse
Affiliation(s)
- Foram Khatsuria
- Centre for Patient Reported Outcomes Research (CPROR), Institute of Applied Health Research, University of Birmingham, Birmingham, UK; NIHR Blood and Transplant Research Unit (BTRU) in Precision Cellular Therapeutics, University of Birmingham, Birmingham, UK; NIHR Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK.
| | - Christel McMullan
- Centre for Patient Reported Outcomes Research (CPROR), Institute of Applied Health Research, University of Birmingham, Birmingham, UK; NIHR Blood and Transplant Research Unit (BTRU) in Precision Cellular Therapeutics, University of Birmingham, Birmingham, UK
| | - Olalekan Lee Aiyegbusi
- Centre for Patient Reported Outcomes Research (CPROR), Institute of Applied Health Research, University of Birmingham, Birmingham, UK; NIHR Blood and Transplant Research Unit (BTRU) in Precision Cellular Therapeutics, University of Birmingham, Birmingham, UK; NIHR Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK; Birmingham Health Partners Centre for Regulatory Science and Innovation, University of Birmingham, Birmingham, UK; NIHR Applied Research Collaboration (ARC) West Midlands, University of Birmingham, Birmingham, UK
| | - Karen L Shaw
- Centre for Patient Reported Outcomes Research (CPROR), Institute of Applied Health Research, University of Birmingham, Birmingham, UK; NIHR Blood and Transplant Research Unit (BTRU) in Precision Cellular Therapeutics, University of Birmingham, Birmingham, UK
| | - Roshina Iqbal
- Centre for Patient Reported Outcomes Research (CPROR), Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Francesca Kinsella
- Birmingham Centre for Cellular Therapy and Transplantation, Birmingham UK
| | - Keith Wilson
- Department of Haematology, Cardiff and Vale University Health Board, Cardiff, UK
| | - Lester Pyatt
- NIHR Blood and Transplant Research Unit (BTRU) in Precision Cellular Therapeutics, Patient Partners, Birmingham, UK
| | - Marlene Lewis
- NIHR Blood and Transplant Research Unit (BTRU) in Precision Cellular Therapeutics, Patient Partners, Birmingham, UK
| | - Sophie M R Wheldon
- NIHR Blood and Transplant Research Unit (BTRU) in Precision Cellular Therapeutics, Patient Partners, Birmingham, UK; Leukaemia Care, Worcester, UK
| | - David Burns
- University Hospitals Birmingham NHS Foundation Trust, Birmingham UK
| | - Ronjon Chakraverty
- NIHR Blood and Transplant Research Unit (BTRU) in Precision Cellular Therapeutics, University of Birmingham, Birmingham, UK; Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Melanie Calvert
- Centre for Patient Reported Outcomes Research (CPROR), Institute of Applied Health Research, University of Birmingham, Birmingham, UK; NIHR Blood and Transplant Research Unit (BTRU) in Precision Cellular Therapeutics, University of Birmingham, Birmingham, UK; NIHR Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK; Birmingham Health Partners Centre for Regulatory Science and Innovation, University of Birmingham, Birmingham, UK; NIHR Applied Research Collaboration (ARC) West Midlands, University of Birmingham, Birmingham, UK
| | - Sarah E Hughes
- Centre for Patient Reported Outcomes Research (CPROR), Institute of Applied Health Research, University of Birmingham, Birmingham, UK; NIHR Blood and Transplant Research Unit (BTRU) in Precision Cellular Therapeutics, University of Birmingham, Birmingham, UK; Birmingham Health Partners Centre for Regulatory Science and Innovation, University of Birmingham, Birmingham, UK; NIHR Applied Research Collaboration (ARC) West Midlands, University of Birmingham, Birmingham, UK
| |
Collapse
|
31
|
Ju A, Choi S, Jeon Y, Kim K. Lymphodepletion in Chimeric Antigen Receptor T-Cell Therapy for Solid Tumors: A Focus on Brain Tumors. Brain Tumor Res Treat 2024; 12:208-220. [PMID: 39542517 PMCID: PMC11570086 DOI: 10.14791/btrt.2024.0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 11/17/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy, which has demonstrated remarkable efficacy in hematologic malignancies, is being extended to the treatment of refractory solid tumors, including brain tumors. Lymphodepletion (LD) is an essential preconditioning process that enhances CAR-T efficacy by promoting CAR-T cell expansion and persistence in the body, and has become a standard regimen for hematologic cancers. Recent clinical results of CAR-T therapy for solid tumors, including brain tumors, have shown that cyclophosphamide/fludarabine-based preconditioning has potential benefits and is gradually becoming adopted in solid tumor CAR-T trials. Furthermore, some CAR-T trials for solid tumors are attempting to develop LD regimens optimized specifically for solid tumors, distinct from the standard LD regimens used in hematologic cancers. In contrast, CAR-T therapy targeting brain tumors frequently employs locoregionally repeated administration in tumors or cerebrospinal fluid, resulting in less frequent use of LD compared to other solid tumors. Nevertheless, several clinical studies suggest that LD may still provide potential benefits for CAR-T expansion and improvement in clinical responses in systemic CAR-T administration. The studies presented in this review suggest that while LD can be beneficial for enhancing CAR-T efficacy, considerations must be made regarding its compatibility with the CAR-T administration route, potential excessive activation based on CAR-T structural characteristics, and target expression in normal organs. Additionally, given the unique characteristics of brain tumors, optimized selection of LD agents, as well as dosing and regimens, may be required, highlighting the need for further research.
Collapse
Affiliation(s)
- Anna Ju
- R&D Center, CellabMED Inc., Seoul, Korea
| | | | | | - Kiwan Kim
- R&D Center, CellabMED Inc., Seoul, Korea.
| |
Collapse
|
32
|
Jawdekar R, Mishra V, Hatgoankar K, Tiwade YR, Bankar NJ. Precision medicine in cancer treatment: Revolutionizing care through proteomics, genomics, and personalized therapies. J Cancer Res Ther 2024; 20:1687-1693. [DOI: 10.4103/jcrt.jcrt_108_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/20/2024] [Indexed: 01/03/2025]
Abstract
ABSTRACT
Recent developments in biotechnology have allowed us to identify unique and complicated biological traits associated with cancer. Genomic profiling through next-generation sequencing (NGS) has revolutionized cancer therapy by evaluating hundreds of genes and biomarkers in a single assay. Proteomics offers blood-based biomarkers for cancer detection, categorization, and therapy monitoring. Immune oncology and chimeric antigen receptor (CAR-T cell) therapy use the immune system to combat cancer. Personalized cancer treatment is on the rise. Although precision medicine holds great promise, its widespread application faces obstacles such as lack of agreement on nomenclature, the difficulty of classifying patients into distinct groups, the difficulties of multimorbidity, magnitude, and the need for prompt intervention. This review studies advances in the era of precision medicine for cancer treatment; the application of genomic profiling techniques, NGS, proteomics, and targeted therapy; and the challenge in the application of precision medicine and the beneficial future it holds in cancer treatment.
Collapse
Affiliation(s)
- Riddhi Jawdekar
- Department of Pathology, Datta Meghe Medical College, Datta Meghe Institute of Higher Education and Research, Nagpur, Maharashtra, India
| | - Vaishnavi Mishra
- Department of Microbiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| | - Kajal Hatgoankar
- Department of Pathology, Datta Meghe Medical College, Datta Meghe Institute of Higher Education and Research, Nagpur, Maharashtra, India
| | - Yugeshwari R. Tiwade
- Department of Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| | - Nandkishor J. Bankar
- Department of Microbiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| |
Collapse
|
33
|
Ye J, Li D, Jie Y, Luo H, Zhang W, Qiu C. Exosome-based nanoparticles and cancer immunotherapy. Biomed Pharmacother 2024; 179:117296. [PMID: 39167842 DOI: 10.1016/j.biopha.2024.117296] [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: 06/20/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
Over the past decades, cancer immunotherapy has encountered challenges such as immunogenicity, inefficiency, and cytotoxicity. Consequently, exosome-based cancer immunotherapy has gained rapid traction as a promising alternative. Exosomes, a type of extracellular vesicles (EVs) ranging from 50 to 150 nm, are self-originating and exhibit fewer side effects compared to traditional therapies. Exosome-based immunotherapy encompasses three significant areas: cancer vaccination, co-inhibitory checkpoints, and adoptive T-cell therapy. Each of these fields leverages the inherent advantages of exosomes, demonstrating substantial potential for individualized tumor therapy and precision medicine. This review aims to elucidate the reasons behind the promise of exosome-based nanoparticles as cancer therapies by examining their characteristics and summarizing the latest research advancements in cancer immunotherapy.
Collapse
Affiliation(s)
- Jiarong Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi Province, 330000 China.
| | - Danni Li
- Second Clinical Medical School, Nanchang University, Jiangxi Province 330000, China
| | - Yiting Jie
- Second Clinical Medical School, Nanchang University, Jiangxi Province 330000, China
| | - Hongliang Luo
- Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, China
| | - Wenjun Zhang
- Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, China
| | - Cheng Qiu
- Gastrointestinal Surgery, Pingxiang People's Hospital, Jiangxi Province 330000, China.
| |
Collapse
|
34
|
Voorhees TJ, Bezerra E, Denlinger N, Jaglowski S, de Lima M. SOHO State of the Art Updates and Next Questions Updates on Building Your CAR-T Cell Program. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:649-652. [PMID: 38643029 DOI: 10.1016/j.clml.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 03/14/2024] [Indexed: 04/22/2024]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has significantly impacted treatment algorithms and clinical outcomes for a variety of patients with hematologic malignancies over the past decade. The field of cellular immunotherapy is currently experiencing a rapid expansion of the number of patients eligible for CAR-T therapies as approvals are being seen in earlier lines of therapy. With the expanded patients eligible for these therapies, more treatment centers will be necessary to keep up with demand. Building a cellular therapy program can be a daunting task, and therefore, we present our experience with building a clinical cellular therapy program.
Collapse
Affiliation(s)
- Timothy J Voorhees
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH.
| | - Evandro Bezerra
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| | - Nathan Denlinger
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| | - Samantha Jaglowski
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| | - Marcos de Lima
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| |
Collapse
|
35
|
Abenavoli EM, Linguanti F, Dercle L, Berti V, Lopci E. FDG-PET/CT Imaging in Chimeric Antigen Receptor-Engineered T-Cell Treatment in Patients with B-Cell Lymphoma: Current Evidence. PET Clin 2024; 19:505-513. [PMID: 38969565 DOI: 10.1016/j.cpet.2024.05.006] [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] [Indexed: 07/07/2024]
Abstract
The Food and Drug Administration and the European Medicines Agency have recently approved chimeric antigen receptor-engineered (CAR) T cells to treat several refractory/relapsed B-cell lymphomas. This comprehensive review aims to demonstrate the pivotal role that [18F]-FDG PET/computed tomographic (CT) imaging can play to enhance the care of patients treated with CAR T-cell therapy. To this end, this review deciphers evidence showing the diagnostic, prognostic, predictive, and theragnostic value of [18F]-FDG PET/CT-derived parameters.
Collapse
Affiliation(s)
| | - Flavia Linguanti
- Nuclear Medicine Department, Ospedale San Donato, Via Pietro Nenni 20, Arezzo 52100, Italy; Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Largo Brambilla 3, Florence 50134, Italy
| | - Laurent Dercle
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, 622 West 168th Street, New York, NY 10032, USA
| | - Valentina Berti
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Largo Brambilla 3, Florence 50134, Italy
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS-Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milano CAP 20089, Italy.
| |
Collapse
|
36
|
Bartoszewska E, Tota M, Kisielewska M, Skowron I, Sebastianka K, Stefaniak O, Molik K, Rubin J, Kraska K, Choromańska A. Overcoming Antigen Escape and T-Cell Exhaustion in CAR-T Therapy for Leukemia. Cells 2024; 13:1596. [PMID: 39329777 PMCID: PMC11430486 DOI: 10.3390/cells13181596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Leukemia is a prevalent pediatric cancer with significant challenges, particularly in relapsed or refractory cases. Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a personalized cancer treatment, modifying patients' T cells to target and destroy resistant cancer cells. This study reviews the current therapeutic options of CAR-T therapy for leukemia, addressing the primary obstacles such as antigen escape and T-cell exhaustion. We explore dual-targeting strategies and their potential to improve treatment outcomes by preventing the loss of target antigens. Additionally, we examine the mechanisms of T-cell exhaustion and strategies to enhance CAR-T persistence and effectiveness. Despite remarkable clinical successes, CAR-T therapy poses risks such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Our findings highlight the need for ongoing research to optimize CAR-T applications, reduce toxicities, and extend this innovative therapy to a broader range of hematologic malignancies. This comprehensive review aims to provide valuable insights for improving leukemia treatment and advancing the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Elżbieta Bartoszewska
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Maciej Tota
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Monika Kisielewska
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Izabela Skowron
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Kamil Sebastianka
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Oliwia Stefaniak
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Klaudia Molik
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Jakub Rubin
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Karolina Kraska
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
37
|
Mohammadi V, Esmaeilzadeh K, Esmaeilzadeh A. Application of magnetic nanoparticles in adoptive cell therapy of cancer; training, guiding and imaging cells. Nanomedicine (Lond) 2024; 19:2315-2329. [PMID: 39258568 PMCID: PMC11488091 DOI: 10.1080/17435889.2024.2395239] [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: 03/24/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024] Open
Abstract
Adoptive cell therapy (ACT) is on the horizon as a thrilling therapeutic plan for cancer. However, widespread application of ACT is often restricted by several challenges, including complexity of priming tumor-specific T cells and poor trafficking in solid tumors. The convergence of nanotechnology and cancer immunotherapy is coming of age and could address the limitations of ACT. Recent studies have provided evidence on the application of magnetic nanoparticles (MNPs) to generate smart immune cells and to bypass problems associated with conventional ACT. Herein, we review current progress in the application of MNPs to improve preparing, guiding and tracking immune cells in cancer ACT. Besides, we comment on the challenges ahead and strategies to optimize MNPs for clinical settings.
Collapse
Affiliation(s)
- Vahid Mohammadi
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kimia Esmaeilzadeh
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
38
|
Armstrong A, Tang Y, Mukherjee N, Zhang N, Huang G. Into the storm: the imbalance in the yin-yang immune response as the commonality of cytokine storm syndromes. Front Immunol 2024; 15:1448201. [PMID: 39318634 PMCID: PMC11420043 DOI: 10.3389/fimmu.2024.1448201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024] Open
Abstract
There is a continuous cycle of activation and contraction in the immune response against pathogens and other threats to human health in life. This intrinsic yin-yang of the immune response ensures that inflammatory processes can be appropriately controlled once that threat has been resolved, preventing unnecessary tissue and organ damage. Various factors may contribute to a state of perpetual immune activation, leading to a failure to undergo immune contraction and development of cytokine storm syndromes. A literature review was performed to consider how the trajectory of the immune response in certain individuals leads to cytokine storm, hyperinflammation, and multiorgan damage seen in cytokine storm syndromes. The goal of this review is to evaluate how underlying factors contribute to cytokine storm syndromes, as well as the symptomatology, pathology, and long-term implications of these conditions. Although the recognition of cytokine storm syndromes allows for universal treatment with steroids, this therapy shows limitations for symptom resolution and survival. By identifying cytokine storm syndromes as a continuum of disease, this will allow for a thorough evaluation of disease pathogenesis, consideration of targeted therapies, and eventual restoration of the balance in the yin-yang immune response.
Collapse
Affiliation(s)
- Amy Armstrong
- Department of Cell Systems and Anatomy, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Microbiology, Immunology, and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Yuting Tang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Neelam Mukherjee
- Department of Microbiology, Immunology, and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Urology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Nu Zhang
- Department of Microbiology, Immunology, and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Gang Huang
- Department of Cell Systems and Anatomy, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Microbiology, Immunology, and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Pathology & Laboratory Medicine, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
39
|
Cyrelle Ornella MS, Kim JJ, Cho E, Cho M, Hwang TH. Dose Considerations for Vaccinia Oncolytic Virus Based on Retrospective Reanalysis of Early and Late Clinical Trials. Vaccines (Basel) 2024; 12:1010. [PMID: 39340040 PMCID: PMC11435715 DOI: 10.3390/vaccines12091010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Over the past decade, oncolytic viruses (OVs) have been developed as a promising treatment alone or in combination in immuno-oncology but have faced challenges in late-stage clinical trials. Our retrospective reanalysis of vaccinia oncolytic virus (VOV) clinical trials indicates that lower doses-rather than the maximum tolerated dose (MTD)-are associated with better tumor response rates. Patients who responded well to lower doses generally had prolonged survival rates in the early phase clinical trial. The association between poor outcomes and an increase in OV-induced neutrophils (OV-N) but not baseline neutrophil counts suggests the need for a comprehensive characterization of OV-N. Although this reanalysis is limited by patient heterogeneity-including differences in cancer type and stage, treatment schedules, and administration routes-it remains informative given the complexities of translational studies in the tumor-bearing mouse models of vaccinia oncolytic viruses. Notably, while OV-N increases with higher viral doses, the immune state shaped by tumor progression likely amplifies this tendency. These findings highlight the importance of OV-N immune modulation as well as dose optimization for the successful clinical development of VOV.
Collapse
Affiliation(s)
- Mefotse Saha Cyrelle Ornella
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam-si 13554, Republic of Korea
| | - Jae-Joon Kim
- Division of Hematology & Oncology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Euna Cho
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam-si 13554, Republic of Korea
| | - Mong Cho
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam-si 13554, Republic of Korea
| | - Tae-Ho Hwang
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam-si 13554, Republic of Korea
- Medical Research Center, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Clinical Pharmacology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| |
Collapse
|
40
|
Gandhi M, Sharma B, Nair S, Vaidya ADB. Current Insights into CAR T-Cell-Based Therapies for Myelodysplastic Syndrome. Pharm Res 2024; 41:1757-1773. [PMID: 39187686 DOI: 10.1007/s11095-024-03761-8] [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: 03/30/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024]
Abstract
Myelodysplastic syndromes (MDS) are due to defective hematopoiesis in bone marrow characterized by cytopenia and dysplasia of blood cells, with a varying degree of risk of acute myeloid leukemia (AML). Currently, the only potentially curative strategy is hematopoietic stem cell transplantation (HSCT). Many patients are ineligible for HSCT, due to late diagnosis, presence of co-morbidities, old age and complications likely due to graft-versus-host disease (GvHD). As a consequence, patients with MDS are often treated conservatively with blood transfusions, chemotherapy, immunotherapy etc. based on the grade and manifestations of MDS. The development of chimeric antigen receptor (CAR)-T cell therapy has revolutionized immunotherapy for hematological malignancies, as evidenced by a large body of literature. However, resistance and toxicity associated with it are also a challenge. Hence, there is an urgent need to develop new strategies for immunological and hematopoetic management of MDS. Herein, we discuss current limitations of CAR T-cell therapy and summarize novel approaches to mitigate this. Further, we discuss the in vivo activation of tumor-specific T cells, immune check inhibitors (ICI) and other approaches to normalize the bone marrow milieu for the management of MDS.
Collapse
Affiliation(s)
- Manav Gandhi
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Bhirisha Sharma
- University of Mumbai, Santa Cruz (East), Mumbai, 400055, India
| | - Sujit Nair
- Viridis Biopharma Pvt. Ltd, Mumbai, 400022, India.
- Phytoveda Pvt. Ltd, Mumbai, 400022, India.
| | - Ashok D B Vaidya
- Kasturba Health Society-Medical Research Centre, Vile Parle (West), Mumbai, 400056, India
| |
Collapse
|
41
|
Kuan CY, Yang IH, Chang CT, Chen ZY, Lin JN, Kuo WT, Lin YY, Yueh A, Lin FH. Enhanced non-viral gene delivery via calcium phosphate/DNA co-precipitates with low-voltage pulse electroporation in NK-92 cells for immunocellular therapy. APL Bioeng 2024; 8:036107. [PMID: 39131207 PMCID: PMC11315581 DOI: 10.1063/5.0198191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024] Open
Abstract
Achieving high cell transfection efficiency is essential for various cell types in numerous disease applications. However, the efficient introduction of genes into natural killer (NK) cells remains a challenge. In this study, we proposed a design strategy for delivering exogenous genes into the NK cell line, NK-92, using a modified non-viral gene transfection method. Calcium phosphate/DNA nanoparticles (pDNA-CaP NPs) were prepared using co-precipitation methods and combined with low-voltage pulse electroporation to facilitate NK-92 transfection. The results demonstrated that the developed pDNA-CaP NPs exhibited a uniform diameter of approximately 393.9 nm, a DNA entrapment efficiency of 65.8%, and a loading capacity of 15.9%. Furthermore, at three days post-transfection, both the transfection efficiency and cell viability of NK-92 were significantly improved compared to standalone plasmid DNA (pDNA) electroporation or solely relying on the endocytosis pathway of pDNA-CaP NPs. This study provides valuable insights into a novel approach that combines calcium phosphate nanoparticles with low-voltage electroporation for gene delivery into NK-92 cells, offering potential advancements in cell therapy.
Collapse
Affiliation(s)
| | - I-Hsuan Yang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli 35053, Taiwan
| | | | - Zhi-Yu Chen
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli 35053, Taiwan
| | - Jhih-Ni Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 49, Fanglan Rd., Taipei 10672, Taiwan
| | - Wei-Ting Kuo
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 49, Fanglan Rd., Taipei 10672, Taiwan
| | | | - Andrew Yueh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli 35053, Taiwan
| | - Feng-Huei Lin
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
42
|
Zhou Z, Mai Y, Zhang G, Wang Y, Sun P, Jing Z, Li Z, Xu Y, Han B, Liu J. Emerging role of immunogenic cell death in cancer immunotherapy: Advancing next-generation CAR-T cell immunotherapy by combination. Cancer Lett 2024; 598:217079. [PMID: 38936505 DOI: 10.1016/j.canlet.2024.217079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Immunogenic cell death (ICD) is a stress-driven form of regulated cell death (RCD) in which dying tumor cells' specific signaling pathways are activated to release damage-associated molecular patterns (DAMPs), leading to the robust anti-tumor immune response as well as a reversal of the tumor immune microenvironment from "cold" to "hot". Chimeric antigen receptor (CAR)-T cell therapy, as a landmark in anti-tumor immunotherapy, plays a formidable role in hematologic malignancies but falls short in solid tumors. The Gordian knot of CAR-T cells for solid tumors includes but is not limited to, tumor antigen heterogeneity or absence, physical and immune barriers of tumors. The combination of ICD induction therapy and CAR-T cell immunotherapy is expected to promote the intensive use of CAR-T cell in solid tumors. In this review, we summarize the characteristics of ICD, stress-responsive mechanism, and the synergistic effect of various ICD-based therapies with CAR-T cells to effectively improve anti-tumor capacity.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yumiao Mai
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Henan Province Key Laboratory of Cardiac Injury and Repair, Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, 450052, China
| | - Yingjie Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Pan Sun
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhaohe Jing
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jian Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
43
|
Yu B, Kang J, Lei H, Li Z, Yang H, Zhang M. Immunotherapy for colorectal cancer. Front Immunol 2024; 15:1433315. [PMID: 39238638 PMCID: PMC11375682 DOI: 10.3389/fimmu.2024.1433315] [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: 05/16/2024] [Accepted: 07/30/2024] [Indexed: 09/07/2024] Open
Abstract
Colorectal cancer is the third most common cancer and the second most lethal cancer in the world. The main cause of the disease is due to dietary and behavioral factors. The treatment of this complex disease is mainly based on traditional treatments, including surgery, radiotherapy, and chemotherapy. Due to its high prevalence and high morbidity, more effective treatments with fewer side effects are urgently needed. In recent years, immunotherapy has become a potential therapeutic alternative and one of the fastest-developing treatments. Immunotherapy inhibits tumor growth by activating or enhancing the immune system to recognize and attack cancer cells. This review presents the latest immunotherapies for immune checkpoint inhibitors, cell therapy, tumor-infiltrating lymphocytes, and oncolytic viruses. Some of these have shown promising results in clinical trials and are used in clinical treatment.
Collapse
Affiliation(s)
- Bing Yu
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| | - Jian Kang
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| | - Hong Lei
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| | - Zhe Li
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| | - Hao Yang
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| | - Meng Zhang
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| |
Collapse
|
44
|
Zarychta J, Kowalczyk A, Marszołek A, Zawitkowska J, Lejman M. Strategies to overcome tumor microenvironment immunosuppressive effect on the functioning of CAR-T cells in high-grade glioma. Ther Adv Med Oncol 2024; 16:17588359241266140. [PMID: 39156126 PMCID: PMC11327996 DOI: 10.1177/17588359241266140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/18/2024] [Indexed: 08/20/2024] Open
Abstract
Despite significant progress in the treatment of some types of cancer, high-grade gliomas (HGGs) remain a significant clinical problem. In the case of glioblastoma (GBM), the most common solid tumor of the central nervous system in adults, the average survival time from diagnosis is only 15-18 months, despite the use of intensive multimodal therapy. Chimeric antigen receptor (CAR)-expressing T cells, which have already been approved by the Food and Drug Administration for use in the treatment of certain hematologic malignancies, are a new, promising therapeutic option. However, the efficacy of CAR-T cells in solid tumors is lower due to the immunosuppressive tumor microenvironment (TME). Reprogramming the immunosuppressive TME toward a pro-inflammatory phenotype therefore seems particularly important because it may allow for increasing the effectiveness of CAR-T cells in the therapy of solid tumors. The following literature review aims to present the results of preclinical studies showing the possibilities of improving the efficacy of CAR-T in the TME of GBM by reprogramming the TME toward a pro-inflammatory phenotype. It may be achievable thanks to the use of CAR-T in a synergistic therapy in combination with oncolytic viruses, radiotherapy, or epigenetic inhibitors, as well as by supporting CAR-T cells crossing of the blood-brain barrier, normalizing impaired angiogenesis in the TME, improving CAR-T effector functions by cytokine signaling or by blocking/knocking out T-cell inhibitors, and modulating the microRNA expression. The use of CAR-T cells modified in this way in synergistic therapy could lead to the longer survival of patients with HGG by inducing an endogenous anti-tumor response.
Collapse
Affiliation(s)
- Julia Zarychta
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Adrian Kowalczyk
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Anna Marszołek
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, Lublin, Poland
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Racławickie 1, Lublin 20-093, Poland
| |
Collapse
|
45
|
Godakhindi V, Tarannum M, Dam SK, Vivero-Escoto JL. Mesoporous Silica Nanoparticles as an Ideal Platform for Cancer Immunotherapy: Recent Advances and Future Directions. Adv Healthc Mater 2024; 13:e2400323. [PMID: 38653190 PMCID: PMC11305940 DOI: 10.1002/adhm.202400323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/10/2024] [Indexed: 04/25/2024]
Abstract
Cancer immunotherapy recently transforms the traditional approaches against various cancer malignancies. Immunotherapy includes systemic and local treatments to enhance immune responses against cancer and involves strategies such as immune checkpoints, cancer vaccines, immune modulatory agents, mimetic antigen-presenting cells, and adoptive cell therapy. Despite promising results, these approaches still suffer from several limitations including lack of precise delivery of immune-modulatory agents to the target cells and off-target toxicity, among others, that can be overcome using nanotechnology. Mesoporous silica nanoparticles (MSNs) are investigated to improve various aspects of cancer immunotherapy attributed to the advantageous structural features of this nanomaterial. MSNs can be engineered to alter their properties such as size, shape, porosity, surface functionality, and adjuvanticity. This review explores the immunological properties of MSNs and the use of MSNs as delivery vehicles for immune-adjuvants, vaccines, and mimetic antigen-presenting cells (APCs). The review also details the current strategies to remodel the tumor microenvironment to positively reciprocate toward the anti-tumor immune cells and the use of MSNs for immunotherapy in combination with other anti-tumor therapies including photodynamic/thermal therapies to enhance the therapeutic effect against cancer. Last, the present demands and future scenarios for the use of MSNs for cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Varsha Godakhindi
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Mubin Tarannum
- Division of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Sudip Kumar Dam
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Juan L Vivero-Escoto
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| |
Collapse
|
46
|
Tchernonog E, Moignet A, Anota A, Bernard S, Bouguet G, Colin F, Rioufol C, Ysebaert L, Gyan E. Health-related quality of life in patients with hematologic malignancies treated with chimeric antigen receptor T-cell therapy: review and current progress. Haematologica 2024; 109:2401-2419. [PMID: 38450528 PMCID: PMC11290540 DOI: 10.3324/haematol.2022.282363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/29/2024] [Indexed: 03/08/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has transformed the care of patients with relapsed/refractory B-cell-derived hematologic malignancies. To date, six CAR T-cell therapies, targeting either CD19 or B-cell maturation antigen, have received regulatory approval. Along with the promising survival benefit, CAR T-cell therapy is associated with potentially life-threatening adverse events, including cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. While clinical trials evaluating CAR T-cell therapy consistently report the incidence of these adverse events, most trials do not collect health-related quality of life (HRQoL) data. As such, the impact of the CAR T-cell therapy process and related adverse events on the physical and psychological well-being of patients remains uncertain. HRQoL and other patient-reported outcome (PRO) assessments in patients with relapsed or refractory hematologic malignancies are of utmost importance, as individuals may have unmet needs and a high demand for tolerable therapy if a cure is not obtained. In addition, it is important to standardize methods of data collection to better assess the impact of CAR T-cell therapy on quality of life, optimize patients' care and costs, and enable comparisons between different studies. We conducted a literature search up to June 2023 to identify the HRQoL tools used in clinical trials and in real-world studies investigating CAR T-cell therapy in patients with lymphomas or leukemias. In the present comprehensive review, we summarize the most commonly used CAR T-cell specific and non-specific HRQoL tools and discuss how the use of HRQoL and other PRO tools may be optimized.
Collapse
Affiliation(s)
| | - Aline Moignet
- Hematology department, Pontchaillou University Hospital, Rennes
| | - Amélie Anota
- Department of Clinical Research and Innovation and Department of Human and Social Sciences, Centre Léon Bérard, Lyon
| | - Sophie Bernard
- Hematology department, Centre Hospitalier de la Côte Basque, Bayonne
| | - Guy Bouguet
- Ensemble Leucémie Lymphomes Espoir (ELLyE), Paris
| | - Fanny Colin
- Hematology department, Pontchaillou University Hospital, Rennes
| | - Catherine Rioufol
- Clinical Oncology Pharmacy Department, University Lyon I, France - EA 3738 CICLY, University Hospital, Lyon
| | - Loïc Ysebaert
- Toulouse Cancer Research Center (CRCT), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France; Clinical Hematology, IUCT Oncopole, Toulouse University Hospital, Toulouse.
| | - Emmanuel Gyan
- Hematology and cell therapy department, University Hospital, Tours, France; Clinical investigation center, INSERM U1415, University Hospital, Tours
| |
Collapse
|
47
|
Hu T, Kumar AR, Luo Y, Tay A. Automating CAR-T Transfection with Micro and Nano-Technologies. SMALL METHODS 2024; 8:e2301300. [PMID: 38054597 DOI: 10.1002/smtd.202301300] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/15/2023] [Indexed: 12/07/2023]
Abstract
Cancer poses a significant health challenge, with traditional treatments like surgery, radiotherapy, and chemotherapy often lacking in cell specificity and long-term curative potential. Chimeric antigen receptor T cell (CAR-T) therapy,utilizing genetically engineered T cells to target cancer cells, is a promising alternative. However, its high cost limits widespread application. CAR-T manufacturing process encompasses three stages: cell isolation and activation, transfection, and expansion.While the first and last stages have straightforward, commercially available automation technologies, the transfection stage lags behind. Current automated transfection relies on viral vectors or bulk electroporation, which have drawbacks such as limited cargo capacity and significant cell disturbance. Conversely, micro and nano-tool methods offer higher throughput and cargo flexibility, yet their automation remains underexplored.In this perspective, the progress in micro and nano-engineering tools for CAR-T transfection followed by a discussion to automate them is described. It is anticipated that this work can inspire the community working on micro and nano transfection techniques to examine how their protocols can be automated to align with the growing interest in automating CAR-T manufacturing.
Collapse
Affiliation(s)
- Tianmu Hu
- Engineering Science Programme, National University of Singapore, Singapore, 117575, Singapore
| | - Arun Rk Kumar
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yikai Luo
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Tissue Engineering Programme, National University of Singapore, Singapore, 117510, Singapore
| |
Collapse
|
48
|
Chitkara A, Sreenivasan S, Yin Y, Rai M, Sadashiv S. Venous Thromboembolism Risk in Hematological Malignancies Post-Chimeric Antigen Receptor T-Cell (CAR-T) Therapy: A Meta-Analysis of Phase 2 and Phase 3 Clinical Trials. Curr Oncol 2024; 31:4338-4345. [PMID: 39195306 PMCID: PMC11352860 DOI: 10.3390/curroncol31080323] [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: 06/22/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Chimeric Antigen Receptor T-cell (CAR-T) therapy uses genetically engineered T-cells with specific binding sites. This therapy allows for tumor specificity and durable treatment responses for patients with hematological malignancies. In this review, we study the risk of venous thromboembolism (VTE) associated with CAR-T therapy. We searched the National Institutes of Health library, Cochrane Library Databases, ClinicalTrials.gov database, and medical literature search engines PubMed and Google Scholar for Phase 2 and Phase 3 drug-efficacy and safety trials to determine the aggregate incidence and risk of VTE treated with CAR-T. Of 1127 search results, nine studies were identified and included in our meta-analysis. Of the 1017 patients who received therapy, 805 patients (79.15%) experienced some degree of CRS, and 122 patients (11.9%) experienced severe CRS (higher than grade 3). Only three out of one thousand and seventeen patients were reported to have experienced venous thromboembolism. Our study did not find a statistically significant association between increased VTE incidence (OR = 0.0005, 95% CI [0.0001, 0.0017]) and CRS/ICANS (p < 0.0001). There was a 0.0050 (95% confidence interval [0.0019, 0.0132]) relative risk for VTE. In our study, we did not find a statistically significantly increased risk of developing VTE despite CRS and underlying malignancy, which have been associated with increased risk of VTE.
Collapse
Affiliation(s)
- Akshit Chitkara
- Department of Internal Medicine, University of California, Riverside, CA 92521, USA;
| | - Sushanth Sreenivasan
- Department of Internal Medicine, Allegheny Health Consortium, Pittsburgh, PA 15222, USA
| | - Yue Yin
- Allegheny Singer Research Institute, Pittsburgh, PA 15212, USA;
| | - Maitreyee Rai
- Division of Hematology and Cellular Therapy, Allegheny Health Network Cancer Institute, Pittsburgh, PA 15224, USA (S.S.)
| | - Santhosh Sadashiv
- Division of Hematology and Cellular Therapy, Allegheny Health Network Cancer Institute, Pittsburgh, PA 15224, USA (S.S.)
| |
Collapse
|
49
|
Li D, Liang T, Hutchins LE, Wolfarth AA, Ferrando-Martinez S, Lee BH, Ho M. rhIL-7-hyFc, a long-acting interleukin-7, improves efficacy of CAR-T cell therapy in solid tumors. J Immunother Cancer 2024; 12:e008989. [PMID: 39043602 PMCID: PMC11268061 DOI: 10.1136/jitc-2024-008989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor T-cell (CAR-T) therapy has achieved remarkable remission in patients with B-cell malignancies. However, its efficacy in treating solid tumors remains limited. Here, we investigated a combination therapy approach using an engineered long-acting interleukin (IL)-7 (rhIL-7-hyFc or NT-I7) and CAR-T cells targeting three antigens, glypican-2 (GPC2), glypican-3 (GPC3), and mesothelin (MSLN), against multiple solid tumor types including liver cancer, neuroblastoma, ovarian cancer, and pancreatic cancer in mice. METHODS CAR-T cells targeting GPC2, GPC3, and MSLN were used in combination with NT-I7 to assess the anticancer activity. Xenograft tumor models, including the liver cancer orthotopic model, were established using NOD scid gamma mice engrafted with cell lines derived from hepatocellular carcinoma, neuroblastoma, ovarian cancer, and pancreatic cancer. The mice were monitored by bioluminescence in vivo tumor imaging and tumor volume measurement using a caliper. Immunophenotyping of CAR-T cells on NT-I7 stimulation was evaluated for memory markers, exhaust markers, and T-cell signaling molecules by flow cytometry and western blotting. RESULTS Compared with the IL-2 combination, preclinical evaluation of NT-I7 exhibited regression of solid tumors via enhanced occupancy of CD4+ CAR-T, improved T-cell expansion, reduced exhaustion markers (programmed cell death protein 1 or PD-1 and lymphocyte-activation gene 3 or LAG-3) expression, and increased generation of stem cell-like memory CAR-T cells. The STAT5 pathway was demonstrated to be downstream of NT-I7 signaling, mediated by increased expression of the IL-7 receptor expression in CAR-T cells. Furthermore, CAR-T cells improved efficacy against tumors with low antigen density when combined with NT-I7 in mice, presenting an avenue for patients with heterogeneous antigenic profiles. CONCLUSION This study provides a rationale for NT-I7 plus CAR-T cell combination therapy for solid tumors in humans.
Collapse
Affiliation(s)
- Dan Li
- National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | | | | | - Mitchell Ho
- National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
50
|
Liu T, Yao W, Sun W, Yuan Y, Liu C, Liu X, Wang X, Jiang H. Components, Formulations, Deliveries, and Combinations of Tumor Vaccines. ACS NANO 2024; 18:18801-18833. [PMID: 38979917 DOI: 10.1021/acsnano.4c05065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Tumor vaccines, an important part of immunotherapy, prevent cancer or kill existing tumor cells by activating or restoring the body's own immune system. Currently, various formulations of tumor vaccines have been developed, including cell vaccines, tumor cell membrane vaccines, tumor DNA vaccines, tumor mRNA vaccines, tumor polypeptide vaccines, virus-vectored tumor vaccines, and tumor-in-situ vaccines. There are also multiple delivery systems for tumor vaccines, such as liposomes, cell membrane vesicles, viruses, exosomes, and emulsions. In addition, to decrease the risk of tumor immune escape and immune tolerance that may exist with a single tumor vaccine, combination therapy of tumor vaccines with radiotherapy, chemotherapy, immune checkpoint inhibitors, cytokines, CAR-T therapy, or photoimmunotherapy is an effective strategy. Given the critical role of tumor vaccines in immunotherapy, here, we look back to the history of tumor vaccines, and we discuss the antigens, adjuvants, formulations, delivery systems, mechanisms, combination therapy, and future directions of tumor vaccines.
Collapse
Affiliation(s)
- Tengfei Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyan Yao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyu Sun
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yihan Yuan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Chen Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| |
Collapse
|