1
|
Önder CE, Moustafa-Oglou M, Schröder SM, Hartkopf AD, Koch A, Seitz CM. Precision Immunotherapy Utilizing Adapter CAR-T Cells (AdCAR-T) in Metastatic Breast Cancer Leads to Target Specific Lysis. Cancers (Basel) 2023; 16:168. [PMID: 38201595 PMCID: PMC10778501 DOI: 10.3390/cancers16010168] [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: 10/31/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
A frequent symptom of metastasized breast cancer (BC) includes the development of malignant pleural effusion (MPE), which contains malignant cells derived from the primary tumor site. The poor prognosis of MPE in metastasized BC indicates the necessity for dependable precision oncology and the importance of models representing the heterogenous nature of metastatic BC. In this study, we cultured MPE-derived metastatic tumor cells from four advanced BC patients using organoid technology. We assessed the expression of tumor-associated antigens on MPE-derived organoid lines by flow cytometry (FC). Based on an individual antigen expression pattern, patient-derived organoids were treated with adapter CAR-T cells (AdCAR-T) and biotinylated monoclonal antibodies targeting CD276, HER2, EGFR, TROP2, or EpCAM. Co-culture assays revealed specific organoid lysis by AdCAR-T depending on individual antigen expression patterns. Our results demonstrate that MPE-derived organoids can serve as a reliable tool for assessing the efficacy of AdCAR-T on metastatic BC in a patient-individualized manner. This approach could potentially be applied in a preclinical setting to instruct therapy decisions. Further, our study demonstrates the feasibility of precision immunotherapy utilizing AdCAR-T to target patient-individualized antigen patterns.
Collapse
Affiliation(s)
- Cansu E. Önder
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany;
| | - Moustafa Moustafa-Oglou
- Department of Pediatric Oncology and Hematology, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Sarah M. Schröder
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Ulm, 89081 Ulm, Germany
- Department of Peptide-Based Immunotherapy, University and University Hospital Tübingen, 72076 Tübingen, Germany
| | - Andreas D. Hartkopf
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - André Koch
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany;
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Christian M. Seitz
- Department of Pediatric Oncology and Hematology, University Hospital Tübingen, 72076 Tübingen, Germany;
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, a Partnership between German Cancer Research Center (DKFZ) and University Hospital Tübingen, 81675 Munich, Germany
| |
Collapse
|
2
|
Keshavarz S, Wall JR, Keshavarz S, Vojoudi E, Jafari-Shakib R. Breast cancer immunotherapy: a comprehensive review. Clin Exp Med 2023; 23:4431-4447. [PMID: 37658246 DOI: 10.1007/s10238-023-01177-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 08/18/2023] [Indexed: 09/03/2023]
Abstract
Cancer remains a major health problem despite numerous new medical interventions that have been introduced in recent years. One of the major choices for cancer therapy is so-called adoptive cell therapy (ACT). ACT can be performed using both innate immune cells, including dendritic cells (DCs), natural killer (NK) cells, and γδ T cells and acquired immune T cells. It has become possible to utilize these cells in both their native and modified states in clinical studies. Because of considerable success in cancer treatment, ACT now plays a role in advanced therapy protocols. Genetic engineering of autologous and allogeneic immune cells (T lymphocytes, NK cells, macrophages, etc.) with chimeric antigen receptors (CAR) is a powerful new tool to target specific antigens on cancer cells. The Food and Drug Administration (FDA) in the US has approved certain CAR-T cells for hematologic malignancies and it is hoped that their use can be extended to incorporate a variety of cells, in particular NK cells. However, the ACT method has some limitations, such as the risk of rejection in allogeneic engrafts. Accordingly, numerous efforts are being made to eliminate or minimize this and other complications. In the present review, we have developed a guide to breast cancer (BC) therapy from conventional therapy, through to cell-based approaches, in particular novel technologies including CAR with emphasis on NK cells as a new and safer candidate in this field as well as the more recent aptamer technology, which can play a major role in BC immunotherapy.
Collapse
Affiliation(s)
- Samaneh Keshavarz
- School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Jack R Wall
- University of Notre Dame Australia, Sydney, Australia
| | - Somayeh Keshavarz
- School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Elham Vojoudi
- Regenerative Medicine, Organ Procurement and Transplantation Multidisciplinary Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Reza Jafari-Shakib
- Department of Immunology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
3
|
Dagar G, Gupta A, Masoodi T, Nisar S, Merhi M, Hashem S, Chauhan R, Dagar M, Mirza S, Bagga P, Kumar R, Akil ASAS, Macha MA, Haris M, Uddin S, Singh M, Bhat AA. Harnessing the potential of CAR-T cell therapy: progress, challenges, and future directions in hematological and solid tumor treatments. J Transl Med 2023; 21:449. [PMID: 37420216 PMCID: PMC10327392 DOI: 10.1186/s12967-023-04292-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
Traditional cancer treatments use nonspecific drugs and monoclonal antibodies to target tumor cells. Chimeric antigen receptor (CAR)-T cell therapy, however, leverages the immune system's T-cells to recognize and attack tumor cells. T-cells are isolated from patients and modified to target tumor-associated antigens. CAR-T therapy has achieved FDA approval for treating blood cancers like B-cell acute lymphoblastic leukemia, large B-cell lymphoma, and multiple myeloma by targeting CD-19 and B-cell maturation antigens. Bi-specific chimeric antigen receptors may contribute to mitigating tumor antigen escape, but their efficacy could be limited in cases where certain tumor cells do not express the targeted antigens. Despite success in blood cancers, CAR-T technology faces challenges in solid tumors, including lack of reliable tumor-associated antigens, hypoxic cores, immunosuppressive tumor environments, enhanced reactive oxygen species, and decreased T-cell infiltration. To overcome these challenges, current research aims to identify reliable tumor-associated antigens and develop cost-effective, tumor microenvironment-specific CAR-T cells. This review covers the evolution of CAR-T therapy against various tumors, including hematological and solid tumors, highlights challenges faced by CAR-T cell therapy, and suggests strategies to overcome these obstacles, such as utilizing single-cell RNA sequencing and artificial intelligence to optimize clinical-grade CAR-T cells.
Collapse
Affiliation(s)
- Gunjan Dagar
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Ashna Gupta
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Tariq Masoodi
- Laboratory of Cancer Immunology and Genetics, Sidra Medicine, Doha, Qatar
| | - Sabah Nisar
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Maysaloun Merhi
- National Center for Cancer Care and Research, Hamad Medical Corporation, 3050, Doha, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
| | - Ravi Chauhan
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Manisha Dagar
- Shiley Eye Institute, University of California San Diego, San Diego, CA, USA
| | - Sameer Mirza
- Department of Chemistry, College of Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Puneet Bagga
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rakesh Kumar
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu and Kashmir, 182320, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Pulwama, Jammu and Kashmir, India
| | - Mohammad Haris
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Shahab Uddin
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar.
| | - Mayank Singh
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar.
| |
Collapse
|
4
|
Faeq MH, Al-Haideri M, Mohammad TAM, Gharebakhshi F, Marofi F, Tahmasebi S, Modaresahmadi S. CAR-modified immune cells as a rapidly evolving approach in the context of cancer immunotherapies. Med Oncol 2023; 40:155. [PMID: 37083979 PMCID: PMC10119530 DOI: 10.1007/s12032-023-02019-4] [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: 03/06/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
Nowadays, one of the main challenges clinicians face is malignancies. Through the progression of technology in recent years, tumor nature and tumor microenvironment (TME) can be better understood. Because of immune system involvement in tumorigenesis and immune cell dysfunction in the tumor microenvironment, clinicians encounter significant challenges in patient treatment and normal function recovery. The tumor microenvironment can stop the development of tumor antigen-specific helper and cytotoxic T cells in the tumor invasion process. Tumors stimulate the production of proinflammatory and immunosuppressive factors and cells that inhibit immune responses. Despite the more successful outcomes, the current cancer therapeutic approaches, including surgery, chemotherapy, and radiotherapy, have not been effective enough for tumor eradication. Hence, developing new treatment strategies such as monoclonal antibodies, adaptive cell therapies, cancer vaccines, checkpoint inhibitors, and cytokines helps improve cancer treatment. Among adoptive cell therapies, the interaction between the immune system and malignancies and using molecular biology led to the development of chimeric antigen receptor (CAR) T cell therapy. CAR-modified immune cells are one of the modern cancer therapeutic methods with encouraging outcomes in most hematological and solid cancers. The current study aimed to discuss the structure, formation, subtypes, and application of CAR immune cells in hematologic malignancies and solid tumors.
Collapse
Affiliation(s)
- Mohammed Hikmat Faeq
- Student of General Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maysoon Al-Haideri
- Department of Physiotherapy, Cihan University-Erbil, Kurdistan Region, Erbil, Iraq
| | - Talar Ahmad Merza Mohammad
- Department of Pharmacology, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Farshad Gharebakhshi
- Department of Radiology, School of Medicine, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Shadan Modaresahmadi
- Department of Immunology and Biotechnology, Texas Tech University Health Siences Center, Abilene, TX, USA
| |
Collapse
|
5
|
El-Tanani M, Al Khatib AO, Al-Najjar BO, Shakya AK, El-Tanani Y, Lee YF, Serrano-Aroca Á, Mishra V, Mishra Y, Aljabali AA, Goyal R, Negi P, Farani MR, Binabaj MM, Gholami A, Binabaj MM, Charbe NB, Tambuwala MM. Cellular and molecular basis of therapeutic approaches to breast cancer. Cell Signal 2023; 101:110492. [PMID: 36241056 DOI: 10.1016/j.cellsig.2022.110492] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022]
Abstract
In recent decades, there has been a significant amount of research into breast cancer, with some important breakthroughs in the treatment of both primary and metastatic breast cancers. It's a well-known fact that treating breast cancer is still a challenging endeavour even though physicians have a fantastic toolset of the latest treatment options at their disposal. Due to limitations of current clinical treatment options, traditional chemotherapeutic drugs, and surgical options are still required to address this condition. In recent years, there have been several developments resulting in a wide range of treatment options. This review article discusses the cellular and molecular foundation of chemotherapeutic drugs, endocrine system-based treatments, biological therapies, gene therapy, and innovative techniques for treating breast cancer.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan; Centre for Cancer Research and Cell Biology, Queen's University Belfast, Grosvenor Road, Belfast BT12 6BJ, Northern Ireland, UK; Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, UK.
| | - Arwa Omar Al Khatib
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Belal O Al-Najjar
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Ashok K Shakya
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Yahia El-Tanani
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Grosvenor Road, Belfast BT12 6BJ, Northern Ireland, UK; Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Yin-Fai Lee
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; School of Life Sciences, Faculty of Science and Engineering, Anglia Ruskin University, Cambridge CB1 1PT, UK; Neuroscience, Psychology & Behaviour, College of Life Sciences, University of Leicester, Leicester LE1 9HN, UK
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Yachana Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid 566, Jordan
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173229, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173229, India
| | - Marzieh Ramezani Farani
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), the Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), 1417614411 Tehran, Iran.
| | - Maryam Moradi Binabaj
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Amir Gholami
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Maryam Moradi Binabaj
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Nitin B Charbe
- Center for pharmacometrics and system pharmacology, department of pharmaceutics, college of pharmacy, University of Florida, FL, USA
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; Neuroscience, Psychology & Behaviour, College of Life Sciences, University of Leicester, Leicester LE1 9HN, UK.
| |
Collapse
|
6
|
Ba H, Dai Z, Zhang Z, Zhang P, Yin B, Wang J, Li Z, Zhou X. Antitumor effect of CAR-T cells targeting transmembrane tumor necrosis factor alpha combined with PD-1 mAb on breast cancers. J Immunother Cancer 2023; 11:e003837. [PMID: 36720496 PMCID: PMC10098269 DOI: 10.1136/jitc-2021-003837] [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: 01/09/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Our previous study showed that transmembrane tumor necrosis factor alpha (tmTNF-α) is overexpressed in primary breast cancers including triple-negative breast cancers (TNBCs). Chimeric antigen receptor engineered-T (CAR-T) cells have been successfully used mainly in B-cell malignancies. METHODS We generated CAR-T cells targeting tmTNF-α but not secreted tumor necrosis factor alpha and assessed the antitumor effect of the CAR-T cells on tmTNF-α-expressing breast cancer cells in vitro and in vivo. RESULTS Our tmTNF-α CAR-T cells showed potent cytotoxicity against tmTNF-α-expressing breast cancer cells but not tmTNF-α-negative tumor cells with increased secretion of interferon gamma (IFN-γ) and interleukin (IL)-2 in vitro. In tmTNF-α-overexpressing TNBC-bearing mice, the tmTNF-α CAR-T therapy induced evident tumor regression, prolonged survival and increased serum concentrations of IFN-γ and IL-2. However, we found thattmTNF-α induced programmed death-ligand 1 (PD-L1) expression through the p38 pathway via TNF receptor (TNFR) and through the NF-κB and AKT pathways via outside-to-inside (reverse) signaling, which might limit the efficacy of the CAR-T cell therapy. Blockage of the PD-L1/programmed death-1 (PD-1) pathway by PD-1 monoclonal antibody significantly enhanced the antitumor effect of the tmTNF-α CAR-T cell therapy in vitro and in vivo, and the combination was effective for antiprimary tumors and had a tendency to increase the antimetastasis effect of the CAR-T cell therapy. CONCLUSION Our findings suggest a potent antitumor efficacy of the tmTNF-α CAR-T cells that can be enhanced by anti-PD-L1/PD-1 because high PD-L1 expression in TNBC was induced by the tmTNF-α signaling, indicating a promising individual therapy for tmTNF-α-positive breast cancers including TNBC.
Collapse
Affiliation(s)
- Hongping Ba
- Department of Immunology, College of Basic Medicine of Tongji Medical College of Huazhong University of Scince and Technology, Wuhan, Hubei, People's Republic of China
| | - Zigang Dai
- Department of Hematology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Zunyue Zhang
- Department of Immunology, College of Basic Medicine of Tongji Medical College of Huazhong University of Scince and Technology, Wuhan, Hubei, People's Republic of China
| | - Peng Zhang
- Department of Immunology, College of Basic Medicine of Tongji Medical College of Huazhong University of Scince and Technology, Wuhan, Hubei, People's Republic of China
| | - Bingjiao Yin
- Department of Immunology, College of Basic Medicine of Tongji Medical College of Huazhong University of Scince and Technology, Wuhan, Hubei, People's Republic of China
| | - Jing Wang
- Department of Immunology, College of Basic Medicine of Tongji Medical College of Huazhong University of Scince and Technology, Wuhan, Hubei, People's Republic of China
| | - Zhuoya Li
- Department of Immunology, College of Basic Medicine of Tongji Medical College of Huazhong University of Scince and Technology, Wuhan, Hubei, People's Republic of China
| | - Xiaoxi Zhou
- Department of Hematology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
An In Vitro Comparison of Costimulatory Domains in Chimeric Antigen Receptor T Cell for Breast Cancer Treatment. J Immunol Res 2022; 2022:2449373. [DOI: 10.1155/2022/2449373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/27/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022] Open
Abstract
Adoptive cellular therapy with chimeric antigen receptor (CAR) T cells has emerged as a potential novel treatment for various cancers. In this study, we have generated CAR T cells targeting mucin-1 (MUC1), which is an aberrantly glycosylated antigen overexpressed on breast cancer cells. Two different signaling domains, including CD28 and 41BB, were incorporated and directly compared the superiority of different costimulatory signals. Two different CAR MUC1 constructs were transduced into primary T cells and evaluated their characteristics and antitumor activities against MUC1+ cancer cells. CAR MUC1 T cells showed high transduction efficiency and antigen specificity toward MUC1+ cancer cell lines and primary breast cancer cells. When coculturing with target cells, the transduced cells exhibited potent antitumor activity in vitro and secrete proinflammatory cytokines. Upon antigen stimulation, incorporation of the 41BB signaling domain was able to improve T cell proliferation and reduce surface PD1 expression and the upregulation of suppressive cytokines, when compared with CAR MUC1 containing the CD28 domain. Our findings show that CAR T cell targeting MUC1 can be effective against MUC1+ breast cancer cell and support the further development of CAR MUC1 T cells containing 41BB signaling in preclinical and clinical studies of breast cancer treatment.
Collapse
|
8
|
Jungles KM, Holcomb EA, Pearson AN, Jungles KR, Bishop CR, Pierce LJ, Green MD, Speers CW. Updates in combined approaches of radiotherapy and immune checkpoint inhibitors for the treatment of breast cancer. Front Oncol 2022; 12:1022542. [PMID: 36387071 PMCID: PMC9643771 DOI: 10.3389/fonc.2022.1022542] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/27/2022] [Indexed: 12/05/2022] Open
Abstract
Breast cancer is the most prevalent non-skin cancer diagnosed in females and developing novel therapeutic strategies to improve patient outcomes is crucial. The immune system plays an integral role in the body’s response to breast cancer and modulating this immune response through immunotherapy is a promising therapeutic option. Although immune checkpoint inhibitors were recently approved for the treatment of breast cancer patients, not all patients respond to immune checkpoint inhibitors as a monotherapy, highlighting the need to better understand the biology underlying patient response. Additionally, as radiotherapy is a critical component of breast cancer treatment, understanding the interplay of radiation and immune checkpoint inhibitors will be vital as recent studies suggest that combined therapies may induce synergistic effects in preclinical models of breast cancer. This review will discuss the mechanisms supporting combined approaches with radiotherapy and immune checkpoint inhibitors for the treatment of breast cancer. Moreover, this review will analyze the current clinical trials examining combined approaches of radiotherapy, immunotherapy, chemotherapy, and targeted therapy. Finally, this review will evaluate data regarding treatment tolerance and potential biomarkers for these emerging therapies aimed at improving breast cancer outcomes.
Collapse
Affiliation(s)
- Kassidy M. Jungles
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States
| | - Erin A. Holcomb
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Ashley N. Pearson
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Kalli R. Jungles
- Department of Biology, Saint Mary’s College, Notre Dame, IN, United States
| | - Caroline R. Bishop
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Lori J. Pierce
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Michael D. Green
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States
- Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
- *Correspondence: Michael D. Green, ; Corey W. Speers,
| | - Corey W. Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Radiation Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Case Comprehensive Cancer Center, Cleveland, OH, United States
- *Correspondence: Michael D. Green, ; Corey W. Speers,
| |
Collapse
|
9
|
Keshavarz A, Salehi A, Khosravi S, Shariati Y, Nasrabadi N, Kahrizi MS, Maghsoodi S, Mardi A, Azizi R, Jamali S, Fotovat F. Recent findings on chimeric antigen receptor (CAR)-engineered immune cell therapy in solid tumors and hematological malignancies. Stem Cell Res Ther 2022; 13:482. [PMID: 36153626 PMCID: PMC9509604 DOI: 10.1186/s13287-022-03163-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 08/12/2022] [Indexed: 11/10/2022] Open
Abstract
Advancements in adoptive cell therapy over the last four decades have revealed various new therapeutic strategies, such as chimeric antigen receptors (CARs), which are dedicated immune cells that are engineered and administered to eliminate cancer cells. In this context, CAR T-cells have shown significant promise in the treatment of hematological malignancies. However, many obstacles limit the efficacy of CAR T-cell therapy in both solid tumors and hematological malignancies. Consequently, CAR-NK and CAR-M cell therapies have recently emerged as novel therapeutic options for addressing the challenges associated with CAR T-cell therapies. Currently, many CAR immune cell trials are underway in various human malignancies around the world to improve antitumor activity and reduce the toxicity of CAR immune cell therapy. This review will describe the comprehensive literature of recent findings on CAR immune cell therapy in a wide range of human malignancies, as well as the challenges that have emerged in recent years.
Collapse
Affiliation(s)
- Ali Keshavarz
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Salehi
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Islamic Azad University,, Isfahan (Khorasgan) Branch, Isfahan, Iran
| | - Setareh Khosravi
- Department of Orthodontics, School of Dentistry, Alborz University of Medical Sciences, Karaj, Iran
| | - Yasaman Shariati
- Department of General Surgery, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Navid Nasrabadi
- Department of Endodontics, School of Dentistry, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Sairan Maghsoodi
- Department of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Amirhossein Mardi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramyar Azizi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Jamali
- Department of Endodontics, College of Stomatology, Stomatological Hospital, Xi’an Jiaotong University, Shaanxi, People’s Republic of China
| | - Farnoush Fotovat
- Department of Prosthodontics, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
10
|
Jie J, Mao D, Cao J, Feng P, Yang P. Customized Multifunctional Peptide Hydrogel Scaffolds for CAR-T-Cell Rapid Proliferation and Solid Tumor Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:37514-37527. [PMID: 35944246 DOI: 10.1021/acsami.2c10727] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
CAR-T-cell therapies must be expanded to obtain a large number of effector cells quickly, and the current technology cannot address this challenge. A longer operational time would lose or alter the function and phenotype of CAR-T cells in response to therapy, and it also causes a loss in the optimal treatment time for patients. At present, lower survival time and homing efficiency reduce the antitumor effect of CAR-T in vivo. But nobody has solved these two issues in one system, which has a similar microenvironment of lymphoid organs to activate/expand cell delivery for immunotherapy. Here, we generated artificial, customized immune cell matrix scaffolds based on a self-assembling peptide to preserve and augment the cell phenotype in light of the characteristics of CAR-T. The all-in-one nanoscale matrix scaffolds reduced the processing time of CAR-T to 3 days and resulted in over a 10-fold increase compared with the traditional protocol. The cells were combined to modulate mechanotransduction and chemical signals, and the mimic matrix scaffolds showed optimal stiffness and adhesive ligand density, thereby accelerating CAR-T-cell proliferation. Meanwhile, engineering CAR-T-secreted intrinsic PD-1 blocking single-chain variable fragments (scFv) further increased cell proliferation and cytotoxicity by resisting the self and tumor microenvironment in a paracrine and autocrine manner. Local delivery of CAR-T cells from the scaffolds significantly enabled long-term retention, suppressed tumor growth, and increased infiltration of effector T cells compared with traditional CAR-T treatment. The application of bioengineering and genetic engineering approaches has led to the development of rapid culture environments that can control matrix scaffold properties for CAR-T-cell and cancer immunotherapies.
Collapse
Affiliation(s)
- Jing Jie
- Department of Clinical Laboratory, The First People's Hospital of Nantong, The Second Affiliated Hospital of Nantong University, 226001 Nantong, P. R. China
| | - Duo Mao
- Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, 510080 Guangzhou, P. R. China
| | - Jie Cao
- Department of Pathology, The First People's Hospital of Nantong, The Second Affiliated Hospital of Nantong University, 226001 Nantong, P. R. China
| | - Panfeng Feng
- Department of Pharmacy, The First People's Hospital of Nantong, The Second Affiliated Hospital of Nantong University, 226001 Nantong, P. R. China
| | - Pengxiang Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, P. R. China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin Medical University, 150081 Harbin, P. R. China
| |
Collapse
|
11
|
Li Z, Yang D, Guo T, Lin M. Advances in MUC1-Mediated Breast Cancer Immunotherapy. Biomolecules 2022; 12:biom12070952. [PMID: 35883508 PMCID: PMC9313386 DOI: 10.3390/biom12070952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/04/2023] Open
Abstract
Breast cancer (BRCA) is the leading cause of death from malignant tumors among women. Fortunately, however, immunotherapy has recently become a prospective BRCA treatment with encouraging achievements and mild safety profiles. Since the overexpression and aberrant glycosylation of MUC1 (human mucin) are closely associated with BRCA, it has become an ideal target for BRCA immunotherapies. In this review, the structure and function of MUC1 are briefly introduced, and the main research achievements in different kinds of MUC1-mediated BRCA immunotherapy are highlighted, from the laboratory to the clinic. Afterward, the future directions of MUC1-mediated BRCA immunotherapy are predicted, addressing, for example, urgent issues in regard to how efficient immunotherapeutic strategies can be generated.
Collapse
Affiliation(s)
- Zhifeng Li
- Medical School of Nantong University, Nantong 226019, China; (Z.L.); (D.Y.)
| | - Dazhuang Yang
- Medical School of Nantong University, Nantong 226019, China; (Z.L.); (D.Y.)
| | - Ting Guo
- Research Center of Clinical Medicine, Jiangsu Taizhou People’s Hospital (Affiliated Hospital 5 of Nantong University), Taizhou 225300, China;
| | - Mei Lin
- Research Center of Clinical Medicine, Jiangsu Taizhou People’s Hospital (Affiliated Hospital 5 of Nantong University), Taizhou 225300, China;
- Correspondence:
| |
Collapse
|
12
|
Luo C, Wang P, He S, Zhu J, Shi Y, Wang J. Progress and Prospect of Immunotherapy for Triple-Negative Breast Cancer. Front Oncol 2022; 12:919072. [PMID: 35795050 PMCID: PMC9251310 DOI: 10.3389/fonc.2022.919072] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/19/2022] [Indexed: 12/19/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer (estimated 2.3 million new cases in 2020) and the leading cause of cancer death (estimated 685,000 deaths in 2020) in women globally. Breast cancers have been categorized into four major molecular subtypes based on the immunohistochemistry (IHC) expression of classic hormone and growth factor receptors including the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), as well as a proliferation marker Ki-67 protein expression. Triple-negative breast cancer (TNBC), a breast cancer subtype lacking ER, PR, and HER2 expression, is associated with a high metastatic potential and poor prognosis. TNBC accounts for approximately only 15%-20% of new breast cancer diagnoses; it is responsible for most breast cancer-related deaths due to the lack of targeted treatment options for this patient population, and currently, systemic chemotherapy, radiation, and surgical excision remain the major treatment modalities for these patients with TNBC. Although breast cancer patients in general do not have a robust response to the immunotherapy, a subset of TNBC has been demonstrated to have high tumor mutation burden and high tumor-infiltrating lymphocytes, resembling the features observed on melanoma or lung cancers, which can benefit from the treatment of immune checkpoint inhibitors (ICIs). Therefore, the immunogenic nature of this aggressive disease has presented an opportunity for the development of TNBC-targeting immunotherapies. The recent US Food and Drug Administration approval of atezolizumab in combination with the chemotherapeutic agent nab-paclitaxel for the treatment of PD-L1-positive unresectable, locally advanced, or metastatic TNBC has led to a new era of immunotherapy in TNBC treatment. In addition, immunotherapy becomes an active research area, both in the cancer biology field and in the oncology field. In this review, we will extend our coverage on recent discoveries in preclinical research and early results in clinical trials from immune molecule-based therapy including cytokines, monoclonal antibodies, antibody-drug conjugates, bi-specific or tri-specific antibodies, ICIs, and neoantigen cancer vaccines; oncolytic virus-based therapies and adoptive immune cell transfer-based therapies including TIL, chimeric antigen receptor-T (CAR-T), CAR-NK, CAR-M, and T-cell receptor-T. In the end, we will list a series of the challenges and opportunities in immunotherapy prospectively and reveal novel technologies such as high-throughput single-cell sequencing and CRISPR gene editing-based screening to generate new knowledges of immunotherapy.
Collapse
Affiliation(s)
- Chenyi Luo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Shenzhen Research Institute of Beijing University of Chinese Medicine, Shenzhen, China
| | - Peipei Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Siqi He
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jingjing Zhu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Shenzhen Research Institute of Beijing University of Chinese Medicine, Shenzhen, China
| | - Jianxun Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Shenzhen Research Institute of Beijing University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
13
|
Xie J, Zou Y, Gao T, Xie L, Tan D, Xie X. Therapeutic Landscape of Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer. Cancer Control 2022; 29:10732748221099230. [PMID: 35499382 PMCID: PMC9067050 DOI: 10.1177/10732748221099230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/30/2022] [Accepted: 04/18/2022] [Indexed: 01/08/2023] Open
Abstract
Human epidermal growth factor receptor 2-positive breast cancer (HER2+BC) is a common malignancy that is prone to recurrence and metastasis in the early stages, resulting in a poor prognosis for patients. Many studies have suggested that targeted therapy promotes clinical outcomes in HER2+BC. With the introduction of trastuzumab in 1998, the prognosis of patients with early HER2+BC has improved significantly. However, owing to obstinate drug resistance and adverse events, the addition of new agents in standardized treatment has become a research hotspot. These promising agents include antibodies, antibody-drug conjugates, tyrosine kinase inhibitors, and anti-HER2 combined therapies. This article provides a brief description of the biology of BC and the expression of HER2, with the aim to provide an overview of the therapeutic landscape of HER2+BC by reviewing research results and introducing the latest evidence to provide a reference for clinical treatment.
Collapse
Affiliation(s)
- Jindong Xie
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yutian Zou
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ting Gao
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Liming Xie
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Duxun Tan
- South China Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Xiaoming Xie
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
14
|
Balachander GM, Kotcherlakota R, Nayak B, Kedaria D, Rangarajan A, Chatterjee K. 3D Tumor Models for Breast Cancer: Whither We Are and What We Need. ACS Biomater Sci Eng 2021; 7:3470-3486. [PMID: 34286955 DOI: 10.1021/acsbiomaterials.1c00230] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Three-dimensional (3D) models have led to a paradigm shift in disease modeling in vitro, particularly for cancer. The past decade has seen a phenomenal increase in the development of 3D models for various types of cancers with a focus on studying stemness, invasive behavior, angiogenesis, and chemoresistance of cancer cells, as well as contributions of its stroma, which has expanded our understanding of these processes. Cancer biology is moving into exploring the emerging hallmarks of cancer, such as inflammation, immune evasion, and reprogramming of energy metabolism. Studies into these emerging concepts have provided novel targets and treatment options such as antitumor immunotherapy. However, 3D models that can investigate the emerging hallmarks are few and underexplored. As commonly used immunocompromised mice and syngenic mice cannot accurately mimic human immunology, stromal interactions, and metabolism and require the use of prohibitively expensive humanized mice, there is tremendous scope to develop authentic 3D tumor models in these areas. Taking the specific case of breast cancer, we discuss the currently available 3D models, their applications to mimic signaling in cancer, tumor-stroma interactions, drug responses, and assessment of drug delivery systems and therapies. We discuss the lacunae in the development of 3D tumor models for the emerging hallmarks of cancer, for lesser-explored forms of breast cancer, and provide insights to develop such models. We discuss how the next generation of 3D models can provide a better mimic of human cancer modeling compared to xenograft models and the scope toward preclinical models and precision medicine.
Collapse
Affiliation(s)
- Gowri Manohari Balachander
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore-560012, India.,Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, MD9-04-11, 2 Medical Drive, Singapore 117593, Singapore
| | - Rajesh Kotcherlakota
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India
| | - Biswadeep Nayak
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India.,Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, United States.,Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore-560012, India
| | - Dhaval Kedaria
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India
| | - Annapoorni Rangarajan
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore-560012, India.,Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore-560012, India
| | - Kaushik Chatterjee
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore-560012, India.,Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India
| |
Collapse
|
15
|
Singh V, Kumar K, Purohit D, Verma R, Pandey P, Bhatia S, Malik V, Mittal V, Rahman MH, Albadrani GM, Arafah MW, El-Demerdash FM, Akhtar MF, Saleem A, Kamel M, Najda A, Abdel-Daim MM, Kaushik D. Exploration of therapeutic applicability and different signaling mechanism of various phytopharmacological agents for treatment of breast cancer. Biomed Pharmacother 2021; 139:111584. [PMID: 34243623 DOI: 10.1016/j.biopha.2021.111584] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Cancer is one of the most dreaded diseases characterized by uncontrolled proliferation of abnormal cells that occurs due to impairment of cell division and apoptosis process. Cancer is categorized into several types on the basis of affected organs and breast cancer (BC) is the most predominant cause of mortality among women. Although, several synthetic and semi-synthetic therapies have been developed for the treatment of BC but they exhibit numerous serious adverse effects therefore; pharmacological agents with fewer/no side effects need to be explored. Plants and phytoconstituents perhaps fulfill the aforementioned requirement and could serve as a potential and alternative therapy for BC treatment. The ongoing biomedical research, clinical trials and number of patents granted have further boosted the acceptance of the plants and plant-derived constituents in the effective treatment of BC. PURPOSE OF STUDY Various treatment strategies such as checkpoint inhibitors, targeting micro RNA, apoptotic pathway, BRCA-1 gene, P53 protein, P13K/Akt/mTOR pathway, notch signaling pathway, hedgehog/gli-1 signaling pathway, poly-ADP ribose polymerase inhibitors, mitogen-activated protein kinase inhibitors etc. are available for BC. In addition to these synthetic and semi-synthetic drug therapies, several natural constituents such as alkaloids, sesquiterpenes, polyphenols, flavonoids and diterpenoids from medicinal plants, vegetables and fruits are reported to possess promising anti-cancer activity. The purpose of the present review is to highlight the various signaling pathways through which plants/herbs show the anti-cancer potential especially against the BC. STUDY DESIGN The literature for the present study was collected from various databases such as Pubmed, Scopus, Chemical Abstracts, Medicinal and aromatic plant abstracts, Web of Science etc. The different patent databases were also reviewed for the anti-cancer (BC) potential of the particular herbs/plants and their formulations. RESULT AND CONCLUSION In this review, we have discussed the number of plants along with their patents of different herbal formulations which are being used for the treatment of BC and other types of cancers. We have also delineated the different signaling mechanisms through which they inhibit the growth of BC cells. In nutshell, we can conclude that large numbers of herbs or their extracts are reported for the treatment of BC. But still, there is further need for research in-depth to translate the use of natural products clinically BC treatment.
Collapse
Affiliation(s)
- Vandana Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Kuldeep Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India; University Institute of Pharmaceutical Sciences (UIPS), Chandigarh University, Mohali, Punjab, India
| | - Deepika Purohit
- Department of Pharmaceutical Sciences, Indira Gandhi University, Rewari 123401, Haryana, India
| | - Ravinder Verma
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Parijat Pandey
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University Haryana, Manesar, Panchgaon, Haryana 122412, India; Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Vinay Malik
- Department of Zoology, Maharshi Dayanand University, Rohtak 124001, India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Md Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
| | - Ghadeer M Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Mohammed W Arafah
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Fatma M El-Demerdash
- Department of Environmental Studies, Institute of Graduate Studies and Research, University of Alexandria, Alexandria, Egypt
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Pakistan
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegtable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland.
| | - Mohamed M Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India.
| |
Collapse
|
16
|
Gharghani MS, Simonian M, Bakhtiari F, Ghaffari MH, Fazli G, Bayat AA, Negahdari B. Chimeric antigen receptor T-cell therapy for breast cancer. Future Oncol 2021; 17:2961-2979. [PMID: 34156280 DOI: 10.2217/fon-2020-1013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
One of the main reasons that researchers pay enormous attention to immunotherapy is that, despite significant advances in conventional therapy approaches, breast cancer remains the leading cause of death from malignant tumors among women. Genetically modifying T cells with chimeric antigen receptors (CAR) is one of the novel methods that has exhibited encouraging activity with relative safety, further urging investigators to develop several CAR T cells to target overexpressed antigens in breast tumors. This article is aimed not only to present such CAR T cells and discuss their remarkable results but also indicates their shortcomings with the hope of achieving possible strategies for improving therapeutic response.
Collapse
Affiliation(s)
- Mighmig Simonian Gharghani
- Department of Animal Science, College of Agriculture, Isfahan University of Technology, Isfahan, 8415683111, Iran
| | - Miganoosh Simonian
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 14177-55469, Iran
| | - Faezeh Bakhtiari
- Department of Laboratory Sciences, Faculty of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, 71348-14336, Iran
| | - Mozhan Haji Ghaffari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 14177-55469, Iran
| | - Ghazaleh Fazli
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Ali Ahmad Bayat
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 14177-55469, Iran
| |
Collapse
|
17
|
Shi W, Qiu Q, Feng Z, Tong Z, Guo W, Zou F, Yue N, Huang W, Qian H. Design, synthesis and immunological evaluation of self-assembled antigenic peptides from dual-antigen targets: a broad-spectrum candidate for an effective antibreast cancer therapy. J Immunother Cancer 2021; 9:jitc-2021-002523. [PMID: 34083420 PMCID: PMC8183215 DOI: 10.1136/jitc-2021-002523] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2021] [Indexed: 12/11/2022] Open
Abstract
Background Considering the narrow immune response spectrum of a single epitope, and the nanoparticles (NPs) as a novel adjuvant can achieve efficient delivery of antigenic peptides safely, a nano-system (denoted as DSPE-PEG-Man@EM-NPs) based on cathepsin B-responsive antigenic peptides was designed and synthesized. Methods Highly affinitive antigenic peptides were delivered by self-assembled NPs, and targeted erythrocyte membranes acted as a peptide carrier to improve antigenic peptides presentation and to strengthen cytotoxic T-cells reaction. Cathepsin B coupling could release antigenic peptides rapidly in dendritic cells. Results Evaluations showed that DSPE-PEG-Man@EM-NPs had obvious inhibitory effects towards both MCF-7 and MDA-MB-231 human breast cancer cell lines. Conclusion Overall, this strategy provides a novel strategy for boosting cytotoxic T lymphocytes response, thereby expanding the adaptation range of tumor antigenic peptides and improving the therapeutic effect of tumor immunotherapy with nanomedicine.
Collapse
Affiliation(s)
- Wei Shi
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Qianqian Qiu
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.,School of Pharmacy, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng Teachers' University, Yancheng 224002, China
| | - Ziying Feng
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Zhenzhen Tong
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Weiwei Guo
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Feng Zou
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Na Yue
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Wenlong Huang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, Jiangsu, China
| | - Hai Qian
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China .,Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, Jiangsu, China
| |
Collapse
|
18
|
Wang Y, Zhong Q, Li Z, Lin Z, Chen H, Wang P. Integrated Profiling Identifies CCNA2 as a Potential Biomarker of Immunotherapy in Breast Cancer. Onco Targets Ther 2021; 14:2433-2448. [PMID: 33859479 PMCID: PMC8043851 DOI: 10.2147/ott.s296373] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/26/2021] [Indexed: 12/24/2022] Open
Abstract
Introduction Breast cancer is the main reason for cancer-related deaths in women and the most common malignant cancer among women. In recent years, immunosuppressive factors have become a new type of treatment for cancer. However, there are no effective biomarkers for breast cancer immunotherapy. Therefore, exploring immune-related biomarkers is presently an important topic in breast cancer. Methods Gene expression profile data of breast cancer from The Cancer Genome Atlas (TCGA) was downloaded. Scale-free gene co-expression networks were built with weighted gene co-expression network analysis. The correlation of genes was performed with Pearson’s correlation values. The potential associations between clinical features and gene sets were studied, and the hub genes were screened out. Gene Ontology and gene set enrichment analysis were used to reveal the function of hub gene in breast cancer. The gene expression profiles of GSE15852, downloaded from the Gene Expression Omnibus database, were used for hub gene verification. In addition, candidate biomarkers expression in breast cancer was studied. Survival analysis was performed using Log rank test and Kaplan–Meier. Immunohistochemistry was used to analyze the expression of CCNA2. Results A total of 6 modules related to immune cell infiltration were identified via the average linkage hierarchical clustering. According to the threshold criteria (module membership >0.9 and gene significance >0.35), a significant module consisting of 13 genes associated with immune cells infiltration were identified as candidate hub genes after performed with the human protein interaction network. And 3 genes with high correlation to clinical traits were identified as hub genes, which were negatively associated with the overall survival. Among them, the expression of CCNA2 was increased in metastatic breast cancer compare with non-metastatic breast cancer, who underwent immunotherapy. Immunohistochemistry results showed that CCNA2 expression in carcinoma tissues was elevated compared with normal control. Discussion CCNA2 identified as a potential immune therapy marker in breast cancer, which were first reported here and deserved further research.
Collapse
Affiliation(s)
- Yichao Wang
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, 318000, People's Republic of China
| | - Qianyi Zhong
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, 318000, People's Republic of China
| | - Zhaoyun Li
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, 318000, People's Republic of China
| | - Zhu Lin
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, 318000, People's Republic of China
| | - Hanjun Chen
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, 318000, People's Republic of China
| | - Pan Wang
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, 318000, People's Republic of China
| |
Collapse
|
19
|
Seyedmirzaei H, Keshavarz-Fathi M, Razi S, Gity M, Rezaei N. Recent progress in immunotherapy of breast cancer targeting the human epidermal growth factor receptor 2 (HER2). J Oncol Pharm Pract 2021; 27:1235-1244. [PMID: 33530866 DOI: 10.1177/1078155221991636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Breast cancer is responsible for most of the cancer-induced deaths in women around the world. The current review will discuss different approaches of targeting HER2, an epidermal growth factor overexpressed in 30% of breast cancer cases. DATA SOURCES We conducted a search on Pubmed and Scopus databases to find studies relevant to HER2+ breast cancers and targeting HER2 as means of immunotherapy. Out of 1043 articles, 105 studies were included in this review. DATA SUMMARY As well as the introduction of HER2 and breast cancer subtypes, we discussed various aspects of HER2-targeting immunotherapy including monoclonal antibodies, Antibody-drug conjugates (ADCs), Chimeric Antigen Receptor (CAR) T-cells and vaccines. CONCLUSIONS Despite several ways of controlling breast cancer, the need to investigate new drugs and approaches seems to be much significant as this cancer still has a heavy burden on people's health and survival.
Collapse
Affiliation(s)
- Homa Seyedmirzaei
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Breast Cancer Association (BrCA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Gity
- Breast Cancer Association (BrCA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Breast Disease Research Center (BDRC), Advanced Imaging Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Luangwattananun P, Junking M, Sujjitjoon J, Wutti-In Y, Poungvarin N, Thuwajit C, Yenchitsomanus PT. Fourth-generation chimeric antigen receptor T cells targeting folate receptor alpha antigen expressed on breast cancer cells for adoptive T cell therapy. Breast Cancer Res Treat 2021; 186:25-36. [PMID: 33389403 DOI: 10.1007/s10549-020-06032-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Treatment of breast cancer (BC) by standard methods is effective in the early stage, but ineffective in the advanced stage of disease. To develop an adoptive T cell therapy for advanced and severe BC, we generated fourth-generation chimeric antigen receptor (CAR) T cells targeting folate receptor alpha antigen (FRα) expressed on BC cells, and preclinically evaluated their anti-BC activities. METHODS The fourth-generation FRα-CAR T cells containing extracellular FRα-specific single-chain variable fragment (scFv) and three intracellular costimulatory domains (CD28, 4-1BB, and CD27) linked to CD3ζ were generated using a lentiviral system, and then were evaluated for their anti-BC activities in two-dimensional and three-dimensional (spheroid) cultures. RESULTS When our fourth-generation FRα-CAR T cells were cocultured with FRα-expressing MDA-MB-231 BC cell line at an effector to target ratio of 20:1, these CAR T cells specifically lysed 88.7 ± 10.6% of the target cells. Interestingly, the cytotoxic lysis of FRα-CAR T cells was more pronounced in target cells with higher surface FRα expression. This specific cytotoxicity of the CAR T cells was not observed when cocultured with FRα-negative MCF10A normal breast-like cell line at the same ratio (34.3 ± 4.7%). When they were cocultured with MDA-MD-231 spheroid, the FRα-CAR T cells exhibited antitumor activity marked with spheroid size reduction and breakage. CONCLUSION This proof-of-concept study thus shows the feasibility of using these fourth-generation FRα-CAR T cells for adoptive T cell therapy in BC.
Collapse
Affiliation(s)
- Piriya Luangwattananun
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.,Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol, University, Bangkok, 10700, Thailand
| | - Mutita Junking
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand. .,Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol, University, Bangkok, 10700, Thailand.
| | - Jatuporn Sujjitjoon
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.,Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol, University, Bangkok, 10700, Thailand
| | - Yupanun Wutti-In
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.,Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Naravat Poungvarin
- Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Chanitra Thuwajit
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.,Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand. .,Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol, University, Bangkok, 10700, Thailand.
| |
Collapse
|
21
|
Shi W, Tong Z, Qiu Q, Yue N, Guo W, Zou F, Zhou D, Li J, Huang W, Qian H. Novel HLA-A2 restricted antigenic peptide derivatives with high affinity for the treatment of breast cancer expressing NY-ESO-1. Bioorg Chem 2020; 103:104138. [PMID: 32745760 DOI: 10.1016/j.bioorg.2020.104138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022]
Abstract
Tumor immunotherapy based on specific tumor antigen has become the focus for breast cancer, and research into cancer/testes antigens (CTA) is progressing. As an important member in the CTA, NY-ESO-1 plays a crucial role in the treatment and prognosis of breast cancer. In this study, we aimed to improve the binding ability to MHC by designing and synthesizing stable NY-ESO-1-derived peptides, based on NetMHC 4.0 webserver (http://www.cbs.dtu.dk/services/NetMHC/) and HLP webserver (http://crdd.osdd.net/raghava/hlp/pep_both.htm). Moreover, after modification of the lead compound, affinity of the peptides to human leukocyte antigen-A2 (HLA-A2) was determined by a flow cytometry and an inverted fluorescence microscope in T2 cells that show high expression of HLA-A2. The results demonstrated that the affinity of peptides II-4 and II-10 to HLA-A2 was significantly better when compared to others (II-Lead, II-1 ~ II-3, II-5 ~ II-9, II-11 ~ II-15). Further studies indicated that II-4 and II-10, especially II-4, significantly promoted the maturation of HLA-A2-positive human peripheral blood-derived dendritic cells (DCs) from morphology and surface markers, the activation of CD8 + T lymphocytes, and the type-specific killing effect on HLA-A2+/NY-ESO-1+ MDA-MB-231 cells. Molecular docking studies suggested a strong interaction between peptide II-4 and HLA-A2, thereby indicating that the II-4 is a promising candidate with antigenic potential in the field of immunotherapy that needs more studies.
Collapse
Affiliation(s)
- Wei Shi
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Zhenzhen Tong
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Qianqian Qiu
- School of Pharmacy, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng Teachers' University, Yancheng 224002, PR China; Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Na Yue
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Weiwei Guo
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Feng Zou
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Daoguang Zhou
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Jiuhui Li
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Wenlong Huang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Hai Qian
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China.
| |
Collapse
|
22
|
Yang V, Gouveia MJ, Santos J, Koksch B, Amorim I, Gärtner F, Vale N. Breast cancer: insights in disease and influence of drug methotrexate. RSC Med Chem 2020; 11:646-664. [PMID: 33479665 PMCID: PMC7578709 DOI: 10.1039/d0md00051e] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
According to the World Health Organization, cancer is one of the leading causes of morbidity and mortality worldwide. The previously estimated 14 million new cases in the year of 2012 are expected to rise, yearly, over the following 2 decades. Among women, breast cancer is the most common one. In 2012, almost 1.7 million people were diagnosed worldwide and half a million died from the disease. Despite having several treatments available, from surgery to chemotherapy, most of these treatments have severe adverse effects. Chemotherapy has a narrow therapeutic window and requires high dosage treatment in patients with advanced-stage cancers and further need innovative treatment strategies. Although methotrexate (MTX) is not a first line drug used against breast cancer, however, it might be valuable to fight the disease. MTX is an effective and cheap drug that might impair malignant growth without irreversible damage to normal tissues. Nevertheless, while MTX does present some disadvantages including poor solubility and low permeability, several strategies are being used to discover and provide novel and effective targeted treatment against breast cancer. In this review, we analyze the chemotherapy of breast cancer and its relationship with drug MTX.
Collapse
Affiliation(s)
- Vítor Yang
- Department of Molecular Pathology and Immunology , Abel Salazar Biomedical Sciences Institute (ICBAS) , University of Porto , Rua de Jorge Viterbo Ferreira, 228 , 4050-313 Porto , Portugal .
- Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal
| | - Maria João Gouveia
- Department of Molecular Pathology and Immunology , Abel Salazar Biomedical Sciences Institute (ICBAS) , University of Porto , Rua de Jorge Viterbo Ferreira, 228 , 4050-313 Porto , Portugal .
- Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal
| | - Joana Santos
- Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal
| | - Beate Koksch
- Department of Chemistry and Biochemistry , Freie Universität Berlin , Takustrasse 3 , 14195 Berlin , Germany
| | - Irina Amorim
- Department of Molecular Pathology and Immunology , Abel Salazar Biomedical Sciences Institute (ICBAS) , University of Porto , Rua de Jorge Viterbo Ferreira, 228 , 4050-313 Porto , Portugal .
- Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) , Rua Júlio Amaral de Carvalho, 45 , 4200-135 Porto , Portugal
| | - Fátima Gärtner
- Department of Molecular Pathology and Immunology , Abel Salazar Biomedical Sciences Institute (ICBAS) , University of Porto , Rua de Jorge Viterbo Ferreira, 228 , 4050-313 Porto , Portugal .
- Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) , Rua Júlio Amaral de Carvalho, 45 , 4200-135 Porto , Portugal
| | - Nuno Vale
- Department of Molecular Pathology and Immunology , Abel Salazar Biomedical Sciences Institute (ICBAS) , University of Porto , Rua de Jorge Viterbo Ferreira, 228 , 4050-313 Porto , Portugal .
- Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) , Rua Júlio Amaral de Carvalho, 45 , 4200-135 Porto , Portugal
| |
Collapse
|
23
|
Pilipow K, Darwich A, Losurdo A. T-cell-based breast cancer immunotherapy. Semin Cancer Biol 2020; 72:90-101. [PMID: 32492452 DOI: 10.1016/j.semcancer.2020.05.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/22/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy has witnessed a new renaissance with the advent of immune checkpoint inhibitors, which reactivate T cells and foster endogenous anti-tumor responses. The excellent results of immunotherapy in the field of melanoma, renal cancer, lung cancer, and other cancer types that have traditionally been known to be immunogenic, rekindled the interest of the oncology community in extending the benefits to all cancers including breast cancer (BC). In this review, we highlight the current state of using T cells as both markers for clinical practice and therapeutic options for BC.
Collapse
Affiliation(s)
- Karolina Pilipow
- Laboratory of Translational Immunology, Italy; Humanitas Clinical and Research Center - IRCCS - Rozzano, MI, Italy
| | - Abbass Darwich
- Laboratory of Mucosal Immunology and Microbiota, Italy; Humanitas Clinical and Research Center - IRCCS - Rozzano, MI, Italy; Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, MI, Italy
| | - Agnese Losurdo
- Laboratory of Translational Immunology, Italy; Medical Oncology and Hematology Unit, Italy; Humanitas Clinical and Research Center - IRCCS - Rozzano, MI, Italy.
| |
Collapse
|
24
|
Mendoza-Reinoso V, McCauley LK, Fournier PG. Contribution of Macrophages and T Cells in Skeletal Metastasis. Cancers (Basel) 2020; 12:E1014. [PMID: 32326073 PMCID: PMC7226332 DOI: 10.3390/cancers12041014] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023] Open
Abstract
Bone is a common site for metastases with a local microenvironment that is highly conducive for tumor establishment and growth. The bone marrow is replete with myeloid and lymphoid linage cells that provide a fertile niche for metastatic cancer cells promoting their survival and growth. Here, we discuss the role of macrophages and T cells in pro- and anti-tumoral mechanisms, their interaction to support cancer cell growth, and their contribution to the development of skeletal metastases. Importantly, immunotherapeutic strategies targeting macrophages and T cells in cancer are also discussed in this review as they represent a great promise for patients suffering from incurable bone metastases.
Collapse
Affiliation(s)
- Veronica Mendoza-Reinoso
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (V.M.-R.); (L.K.M.)
| | - Laurie K. McCauley
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (V.M.-R.); (L.K.M.)
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Pierrick G.J. Fournier
- Biomedical Innovation Department, Centro de Investigación Científica y de Educación Superior de Ensenada, Ensenada, BC 22860, Mexico
| |
Collapse
|
25
|
Idrissou M, Judes G, Daures M, Sanchez A, El Ouardi D, Besse S, Degoul F, Penault-Llorca F, Bignon YJ, Bernard-Gallon D. TIP60 Inhibitor TH1834 Reduces Breast Cancer Progression in Xenografts in Mice. ACTA ACUST UNITED AC 2019; 23:457-459. [DOI: 10.1089/omi.2019.0126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Mouhamed Idrissou
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand, France
- INSERM—UMR 1240—Molecular Imaging and Theranostic Strategies (IMOST), Clermont-Ferrand, France
| | - Gaëlle Judes
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand, France
- INSERM—UMR 1240—Molecular Imaging and Theranostic Strategies (IMOST), Clermont-Ferrand, France
| | - Marine Daures
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand, France
- INSERM—UMR 1240—Molecular Imaging and Theranostic Strategies (IMOST), Clermont-Ferrand, France
| | - Anna Sanchez
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand, France
- INSERM—UMR 1240—Molecular Imaging and Theranostic Strategies (IMOST), Clermont-Ferrand, France
| | - Driss El Ouardi
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand, France
- INSERM—UMR 1240—Molecular Imaging and Theranostic Strategies (IMOST), Clermont-Ferrand, France
| | - Sophie Besse
- INSERM—UMR 1240—Molecular Imaging and Theranostic Strategies (IMOST), Clermont-Ferrand, France
| | - Françoise Degoul
- INSERM—UMR 1240—Molecular Imaging and Theranostic Strategies (IMOST), Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- INSERM—UMR 1240—Molecular Imaging and Theranostic Strategies (IMOST), Clermont-Ferrand, France
- Department of Biopathology, Centre Jean Perrin, Clermont-Ferrand, France
| | - Yves-Jean Bignon
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand, France
- INSERM—UMR 1240—Molecular Imaging and Theranostic Strategies (IMOST), Clermont-Ferrand, France
| | - Dominique Bernard-Gallon
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand, France
- INSERM—UMR 1240—Molecular Imaging and Theranostic Strategies (IMOST), Clermont-Ferrand, France
| |
Collapse
|
26
|
Zhang Z, Sun L, Zhang Y, Lu G, Li Y, Wei Z. Long non‐coding RNA FEZF1‐AS1 promotes breast cancer stemness and tumorigenesis via targeting miR‐30a/Nanog axis. J Cell Physiol 2018; 233:8630-8638. [DOI: 10.1002/jcp.26611] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/22/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Zhi Zhang
- Department of AnesthesiologyHuaihe HospitalHenan UniversityKaifengChina
| | - Liwei Sun
- Department of AnesthesiologyHuaihe HospitalHenan UniversityKaifengChina
| | - Yixuan Zhang
- Department of AnesthesiologyHuaihe HospitalHenan UniversityKaifengChina
| | - Guanming Lu
- Basic Medical SchoolHenan UniversityKaifengChina
| | - Yongqiang Li
- Basic Medical SchoolHenan UniversityKaifengChina
| | | |
Collapse
|