1
|
Adil S, Banday MT, Wani MA, Hussain SA, Shah S, Sheikh ID, Shafi M, Khan AA, Kashoo ZA, Pattoo RA, Swelum AA. Nano-protected form of rosemary essential oil has a positive influence on blood biochemistry parameters, haematological indices, immune-antioxidant status, intestinal microbiota and histomorphology of meat-type chickens. Poult Sci 2024; 103:104309. [PMID: 39303351 PMCID: PMC11426142 DOI: 10.1016/j.psj.2024.104309] [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: 07/05/2024] [Revised: 08/23/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024] Open
Abstract
This study aimed to investigate the potential of free and nanoprotected rosemary essential oil (REO) as an antibiotic alternative in blood biochemistry, hematology, immune-antioxidant status, intestinal microbiology, and histomorphology of broilers. A total of 420 1-wk-old broiler chicks (Cobb) were randomly allotted into 7 treatments, each having 4 replicates of 15 chicks. The first group served as control received basal diet, while the second group was fed a basal diet plus 100 mg/kg enramycin and third group basal diet plus 150 mg/kg chitosan nanoparticles (CNPs). The fourth and fifth groups received diets supplemented with 100 mg and 200 mg free REO (F-REO)/kg diet. The sixth and seventh groups were supplemented with 100 mg and 200 mg nanoprotected REO (N-REO)/kg diet. Results revealed that supplementing nanoprotected REO significantly (P < 0.05) decreased the levels of blood cholesterol and low-density lipoproteins (LDL) compared to control and enramycin groups. The kidney and liver function parameters were not altered by adding free or nanoprotected REO to the diet. Both levels of nanoprotected REO significantly (P < 0.05) reduced heterophil: lymphocyte (H: L) ratio compared to all other groups. Birds receiving nanoprotected REO at 200 mg/kg diet had significantly (P ˂ 0.05) raised serum levels of immunoglobulin G (IgG) and immunoglobulin M (IgM) compared to control and other birds. Anti-SRBC titre and cell-mediated immunity improved significantly (P < 0.05) in nanoprotected REO groups. Supplementation of nanoprotected REO resulted in significantly (P < 0.05) higher values for superoxide dismutase (SOD), glutathione (GSH) and total antioxidant status (TAS). The caecal microbiota was improved in broiler birds fed diets supplemented with nanoprotected REO. The 200 mg nanoprotected REO/kg diet supplementation resulted in significantly (P < 0.05) better villus height (VH) and villus height: crypt depth (VH: CD) ratio in all the segments of the small intestine. In conclusion, feeding REO in nanoprotected form in a 200 mg/kg diet could be used as an antibiotic substitute to improve broiler chicken's lipid profile, immune-antioxidant status, and intestinal health.
Collapse
Affiliation(s)
- Sheikh Adil
- Division of Livestock Production and Management, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-K 190025, Jammu and Kashmir, India
| | - Muhammad T Banday
- Division of Livestock Production and Management, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-K 190025, Jammu and Kashmir, India
| | - Manzoor A Wani
- Division of Livestock Production and Management, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-K 190025, Jammu and Kashmir, India
| | | | - Showkat Shah
- Division of Veterinary Pathology, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-K 190001, Jammu and Kashmir, India
| | - Islam D Sheikh
- Division of Livestock Production and Management, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-K 190025, Jammu and Kashmir, India
| | - Majid Shafi
- Division of Veterinary Pathology, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-K 190001, Jammu and Kashmir, India
| | - Azmat A Khan
- Division of Livestock Production and Management, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-K 190025, Jammu and Kashmir, India
| | - Zahid A Kashoo
- Division of Veterinary Microbiology, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-K 190001, Jammu and Kashmir, India
| | - Roof A Pattoo
- Division of Livestock Production and Management, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-K 190025, Jammu and Kashmir, India
| | - Ayman A Swelum
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
2
|
Li Y, Chen S, Liu S. Immune Effect of Co-Culture of Dendritic Cells and Cytokine-Induced Killer Cells on Prostate Cancer Cells. Cell Biochem Biophys 2024:10.1007/s12013-024-01569-2. [PMID: 39448420 DOI: 10.1007/s12013-024-01569-2] [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: 09/18/2024] [Indexed: 10/26/2024]
Abstract
It was to explore the immune outcome of co-culture of dendritic cells (DC) and cytokine-induced killer cells (CIK) on prostate cancer (PCa) cells. Peripheral blood mononuclear cells (PBMCs) were extracted from healthy blood donors. DC and CIK cells were induced and co-cultured. The proliferation and phenotypic changes of DC, CIK, and DC-CIK cells/groups were observed. Model rats were constructed by injecting PC3 PCa cells into the abdominal cavity. The successful 50 cases were divided into a negative control group, a chemotherapy group, a DC group, a CIK group, and a DC-CIK treatment group (each consisting of 10 rats) to observe tumor progression. The secreted concentrations of interleukin-12 (IL-12) ((103.67 ± 2.77) pg/mL) and interferon-γ (IFN-γ) ((730.09 ± 23.52) pg/mL) were higher in DC-CIK group as against DC and CIK groups; the proliferation of CIK was higher in DC-CIK group as against CIK within 12 to 20 days of culture. The positive rate (PR) of CD3+/ CD56+ and CD8+ was higher and that of CD45RA+ was lower in DC-CIK group as against CIK.The killing rate of the DC-CIK group was higher than that of the DC and CIK groups at a target effect ratio of 10:1/20:1/50:1 (P < 0.05). After the treatment, the body weight of rats in the chemotherapy group, DC group, and CIK group was significantly reduced (P < 0.05), while no significant changes were observed in the negative control group and DC-CIK group (P > 0.05). After 25 days of treatment, the tumor size in the DC-CIK group was significantly smaller compared to the negative control group, chemotherapy group, DC group, and CIK group; the necrotic area of the tumor tissue in the DC-CIK group was also significantly larger than that in the negative control group, chemotherapy group, DC group, and CIK group (P < 0.05). Co-culture of DC and CIK is excellent in enhancing the proliferation of CIK cells, increasing the secretion of IL-12 and IFN-γ, and enhancing the activity of immune cells and anti-tumor ability, showing its potential in anti-PCa tumor immunotherapy.
Collapse
Affiliation(s)
- Yaojun Li
- Department of Urology Surgery, Tongxiang First People's Hospital, Tongxiang, 314500, Zhejiang, China
| | - Shanmiao Chen
- Department of Urology Surgery, Tongxiang First People's Hospital, Tongxiang, 314500, Zhejiang, China
| | - Shoulei Liu
- Department of Urology Surgery, Tongxiang First People's Hospital, Tongxiang, 314500, Zhejiang, China.
| |
Collapse
|
3
|
Li X, Zhu Y, Yi J, Deng Y, Lei B, Ren H. Adoptive cell immunotherapy for breast cancer: harnessing the power of immune cells. J Leukoc Biol 2024; 115:866-881. [PMID: 37949484 DOI: 10.1093/jleuko/qiad144] [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: 08/15/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023] Open
Abstract
Breast cancer is the most prevalent malignant neoplasm worldwide, necessitating the development of novel therapeutic strategies owing to the limitations posed by conventional treatment modalities. Immunotherapy is an innovative approach that has demonstrated significant efficacy in modulating a patient's innate immune system to combat tumor cells. In the era of precision medicine, adoptive immunotherapy for breast cancer has garnered widespread attention as an emerging treatment strategy, primarily encompassing cellular therapies such as tumor-infiltrating lymphocyte therapy, chimeric antigen receptor T/natural killer/M cell therapy, T cell receptor gene-engineered T cell therapy, lymphokine-activated killer cell therapy, cytokine-induced killer cell therapy, natural killer cell therapy, and γδ T cell therapy, among others. This treatment paradigm is based on the principles of immune memory and antigen specificity, involving the collection, processing, and expansion of the patient's immune cells, followed by their reintroduction into the patient's body to activate the immune system and prevent tumor recurrence and metastasis. Currently, multiple clinical trials are assessing the feasibility, effectiveness, and safety of adoptive immunotherapy in breast cancer. However, this therapeutic approach faces challenges associated with tumor heterogeneity, immune evasion, and treatment safety. This review comprehensively summarizes the latest advancements in adoptive immunotherapy for breast cancer and discusses future research directions and prospects, offering valuable guidance and insights into breast cancer immunotherapy.
Collapse
Affiliation(s)
- Xue Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150076, Heilongjiang, China
| | - Yunan Zhu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150076, Heilongjiang, China
| | - Jinfeng Yi
- Department of Pathology, Harbin Medical University, 157 Baojian Road, Harbin 150081, Heilongjiang, China
| | - Yuhan Deng
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150076, Heilongjiang, China
| | - Bo Lei
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150076, Heilongjiang, China
| | - He Ren
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150076, Heilongjiang, China
| |
Collapse
|
4
|
Pan QZ, Zhao JJ, Liu L, Zhang DS, Wang LP, Hu WW, Weng DS, Xu X, Li YZ, Tang Y, Zhang WH, Li JY, Zheng X, Wang QJ, Li YQ, Xiang T, Zhou L, Yang SN, Wu C, Huang RX, He J, Du WJ, Chen LJ, Wu YN, Xu B, Shen Q, Zhang Y, Jiang JT, Ren XB, Xia JC. XELOX (capecitabine plus oxaliplatin) plus bevacizumab (anti-VEGF-A antibody) with or without adoptive cell immunotherapy in the treatment of patients with previously untreated metastatic colorectal cancer: a multicenter, open-label, randomized, controlled, phase 3 trial. Signal Transduct Target Ther 2024; 9:79. [PMID: 38565886 PMCID: PMC10987514 DOI: 10.1038/s41392-024-01788-2] [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: 09/11/2023] [Revised: 01/30/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Fluoropyrimidine-based combination chemotherapy plus targeted therapy is the standard initial treatment for unresectable metastatic colorectal cancer (mCRC), but the prognosis remains poor. This phase 3 trial (ClinicalTrials.gov: NCT03950154) assessed the efficacy and adverse events (AEs) of the combination of PD-1 blockade-activated DC-CIK (PD1-T) cells with XELOX plus bevacizumab as a first-line therapy in patients with mCRC. A total of 202 participants were enrolled and randomly assigned in a 1:1 ratio to receive either first-line XELOX plus bevacizumab (the control group, n = 102) or the same regimen plus autologous PD1-T cell immunotherapy (the immunotherapy group, n = 100) every 21 days for up to 6 cycles, followed by maintenance treatment with capecitabine and bevacizumab. The main endpoint of the trial was progression-free survival (PFS). The median follow-up was 19.5 months. Median PFS was 14.8 months (95% CI, 11.6-18.0) for the immunotherapy group compared with 9.9 months (8.0-11.8) for the control group (hazard ratio [HR], 0.60 [95% CI, 0.40-0.88]; p = 0.009). Median overall survival (OS) was not reached for the immunotherapy group and 25.6 months (95% CI, 18.3-32.8) for the control group (HR, 0.57 [95% CI, 0.33-0.98]; p = 0.043). Grade 3 or higher AEs occurred in 20.0% of patients in the immunotherapy group and 23.5% in the control groups, with no toxicity-associated deaths reported. The addition of PD1-T cells to first-line XELOX plus bevacizumab demonstrates significant clinical improvement of PFS and OS with well tolerability in patients with previously untreated mCRC.
Collapse
Affiliation(s)
- Qiu-Zhong Pan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Jing-Jing Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Liang Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, PR China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, PR China
- Department of Biotherapy/Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Dong-Sheng Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Li-Ping Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Wen-Wei Hu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - De-Sheng Weng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Xiang Xu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, PR China
| | - Yi-Zhuo Li
- Imaging Diagnosis and Interventional Center, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Yan Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Wei-Hong Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, PR China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, PR China
- Department of Biotherapy/Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Jie-Yao Li
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - Qi-Jing Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Yong-Qiang Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Tong Xiang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Li Zhou
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, PR China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, PR China
- Department of Biotherapy/Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Shuang-Ning Yang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Chen Wu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - Rong-Xing Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Jia He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Wei-Jiao Du
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, PR China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, PR China
- Department of Biotherapy/Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Lu-Jun Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - Yue-Na Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Bin Xu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - Qiong Shen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - Yi Zhang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Jing-Ting Jiang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China.
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China.
| | - Xiu-Bao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, PR China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, PR China.
- Department of Biotherapy/Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China.
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China.
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China.
| |
Collapse
|
5
|
Mei J, Liu X, Tian H, Chen Y, Cao Y, Zeng J, Liu Y, Chen Y, Gao Y, Yin J, Wang P. Tumour organoids and assembloids: Patient-derived cancer avatars for immunotherapy. Clin Transl Med 2024; 14:e1656. [PMID: 38664597 PMCID: PMC11045561 DOI: 10.1002/ctm2.1656] [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: 09/27/2023] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Organoid technology is an emerging and rapidly growing field that shows promise in studying organ development and screening therapeutic regimens. Although organoids have been proposed for a decade, concerns exist, including batch-to-batch variations, lack of the native microenvironment and clinical applicability. MAIN BODY The concept of organoids has derived patient-derived tumour organoids (PDTOs) for personalized drug screening and new drug discovery, mitigating the risks of medication misuse. The greater the similarity between the PDTOs and the primary tumours, the more influential the model will be. Recently, 'tumour assembloids' inspired by cell-coculture technology have attracted attention to complement the current PDTO technology. High-quality PDTOs must reassemble critical components, including multiple cell types, tumour matrix, paracrine factors, angiogenesis and microorganisms. This review begins with a brief overview of the history of organoids and PDTOs, followed by the current approaches for generating PDTOs and tumour assembloids. Personalized drug screening has been practised; however, it remains unclear whether PDTOs can predict immunotherapies, including immune drugs (e.g. immune checkpoint inhibitors) and immune cells (e.g. tumour-infiltrating lymphocyte, T cell receptor-engineered T cell and chimeric antigen receptor-T cell). PDTOs, as cancer avatars of the patients, can be expanded and stored to form a biobank. CONCLUSION Fundamental research and clinical trials are ongoing, and the intention is to use these models to replace animals. Pre-clinical immunotherapy screening using PDTOs will be beneficial to cancer patients. KEY POINTS The current PDTO models have not yet constructed key cellular and non-cellular components. PDTOs should be expandable and editable. PDTOs are promising preclinical models for immunotherapy unless mature PDTOs can be established. PDTO biobanks with consensual standards are urgently needed.
Collapse
Affiliation(s)
- Jie Mei
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of AgingWenzhou Medical UniversityWenzhouPeople's Republic of China
- Department of Clinical Pharmacology, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of PharmacogeneticsCentral South UniversityChangshaPeople's Republic of China
- Engineering Research Center of Applied Technology of PharmacogenomicsMinistry of EducationChangshaPeople's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Xingjian Liu
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of AgingWenzhou Medical UniversityWenzhouPeople's Republic of China
| | - Hui‐Xiang Tian
- Department of Clinical Pharmacology, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of PharmacogeneticsCentral South UniversityChangshaPeople's Republic of China
| | - Yixuan Chen
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of AgingWenzhou Medical UniversityWenzhouPeople's Republic of China
| | - Yang Cao
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of AgingWenzhou Medical UniversityWenzhouPeople's Republic of China
| | - Jun Zeng
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of AgingWenzhou Medical UniversityWenzhouPeople's Republic of China
- Department of Thoracic Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Yung‐Chiang Liu
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of AgingWenzhou Medical UniversityWenzhouPeople's Republic of China
| | - Yaping Chen
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of AgingWenzhou Medical UniversityWenzhouPeople's Republic of China
| | - Yang Gao
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
- Department of Thoracic Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis and Treatment, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
- Xiangya Lung Cancer Center, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Ji‐Ye Yin
- Department of Clinical Pharmacology, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of PharmacogeneticsCentral South UniversityChangshaPeople's Republic of China
- Engineering Research Center of Applied Technology of PharmacogenomicsMinistry of EducationChangshaPeople's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Peng‐Yuan Wang
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of AgingWenzhou Medical UniversityWenzhouPeople's Republic of China
| |
Collapse
|
6
|
Ying Li CM, Li R, Drew P, Price T, Smith E, Maddern GJ, Tomita Y, Fenix K. Clinical application of cytokine-induced killer (CIK) cell therapy in colorectal cancer: Current strategies and future challenges. Cancer Treat Rev 2024; 122:102665. [PMID: 38091655 DOI: 10.1016/j.ctrv.2023.102665] [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: 10/27/2023] [Revised: 11/23/2023] [Accepted: 11/26/2023] [Indexed: 01/01/2024]
Abstract
Colorectal cancer (CRC) remains a significant global health burden and is the second leading cause of cancer-related death. Cytokine induced killer (CIK) cell therapy is an immunotherapy which has the potential to meet this need. Clinical trials of CIK cell therapy for the management of CRC have reported improved clinical outcomes. However, production and delivery protocols varied significantly, and many studies were reported only in Chinese language journals. Here we present the most comprehensive review of the clinical CIK cell therapy trials for CRC management to date. We accessed both English and Chinese language clinical studies, and summarise how CIK cell therapy has been implemented, from manufacturing to patient delivery. We discuss current challenges that impede wider adoption of CIK cell therapy in CRC management.
Collapse
Affiliation(s)
- Celine Man Ying Li
- Department of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia
| | - Runhao Li
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia; Medical Oncology, The Queen Elizabeth Hospital and The University of Adelaide, Woodville, SA 5011, Australia
| | - Paul Drew
- Department of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia
| | - Timothy Price
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia; Medical Oncology, The Queen Elizabeth Hospital and The University of Adelaide, Woodville, SA 5011, Australia
| | - Eric Smith
- Department of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia; Medical Oncology, The Queen Elizabeth Hospital and The University of Adelaide, Woodville, SA 5011, Australia
| | - Guy J Maddern
- Department of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia
| | - Yoko Tomita
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia; Medical Oncology, The Queen Elizabeth Hospital and The University of Adelaide, Woodville, SA 5011, Australia
| | - Kevin Fenix
- Department of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia.
| |
Collapse
|
7
|
Chen Z, Hu Y, Mei H. Advances in CAR-Engineered Immune Cell Generation: Engineering Approaches and Sourcing Strategies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303215. [PMID: 37906032 PMCID: PMC10724421 DOI: 10.1002/advs.202303215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/03/2023] [Indexed: 11/02/2023]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a highly efficacious treatment modality for refractory and relapsed hematopoietic malignancies in recent years. Furthermore, CAR technologies for cancer immunotherapy have expanded from CAR-T to CAR-natural killer cell (CAR-NK), CAR-cytokine-induced killer cell (CAR-CIK), and CAR-macrophage (CAR-MΦ) therapy. Nevertheless, the high cost and complex manufacturing processes of ex vivo generation of autologous CAR products have hampered broader application. There is an urgent need to develop an efficient and economical paradigm shift for exploring new sourcing strategies and engineering approaches toward generating CAR-engineered immune cells to benefit cancer patients. Currently, researchers are actively investigating various strategies to optimize the preparation and sourcing of these potent immunotherapeutic agents. In this work, the latest research progress is summarized. Perspectives on the future of CAR-engineered immune cell manufacturing are provided, and the engineering approaches, and diverse sources used for their development are focused upon.
Collapse
Affiliation(s)
- Zhaozhao Chen
- Institute of HematologyUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic DiseaseWuhan430022China
| | - Yu Hu
- Institute of HematologyUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic DiseaseWuhan430022China
| | - Heng Mei
- Institute of HematologyUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic DiseaseWuhan430022China
| |
Collapse
|
8
|
Cappuzzello E, Vigolo E, D’Accardio G, Astori G, Rosato A, Sommaggio R. How can Cytokine-induced killer cells overcome CAR-T cell limits. Front Immunol 2023; 14:1229540. [PMID: 37675107 PMCID: PMC10477668 DOI: 10.3389/fimmu.2023.1229540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/02/2023] [Indexed: 09/08/2023] Open
Abstract
The successful treatment of patients affected by B-cell malignancies with Chimeric Antigen Receptor (CAR)-T cells represented a breakthrough in the field of adoptive cell therapy (ACT). However, CAR-T therapy is not an option for every patient, and several needs remain unmet. In particular, the production of CAR-T cells is expensive, labor-intensive and logistically challenging; additionally, the toxicities deriving from CAR-T cells infusion, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), have been documented extensively. Alternative cellular therapy products such as Cytokine-induced killer (CIK) cells have the potential to overcome some of these obstacles. CIK cells are a heterogeneous population of polyclonal CD3+CD56+ T cells with phenotypic and functional properties of NK cells. CIK cell cytotoxicity is exerted in a major histocompatibility complex (MHC)-unrestricted manner through the engagement of natural killer group 2 member D (NKG2D) molecules, against a wide range of hematological and solid tumors without the need for prior antigen exposure or priming. The foremost potential of CIK cells lies in the very limited ability to induce graft-versus-host disease (GvHD) reactions in the allogeneic setting. CIK cells are produced with a simple and extremely efficient expansion protocol, which leads to a massive expansion of effector cells and requires a lower financial commitment compared to CAR-T cells. Indeed, CAR-T manufacturing involves the engineering with expensive GMP-grade viral vectors in centralized manufacturing facilities, whereas CIK cell production is successfully performed in local academic GMP facilities, and CIK cell treatment is now licensed in many countries. Moreover, the toxicities observed for CAR-T cells are not present in CIK cell-treated patients, thus further reducing the costs associated with hospitalization and post-infusion monitoring of patients, and ultimately encouraging the delivery of cell therapies in the outpatient setting. This review aims to give an overview of the limitations of CAR-T cell therapy and outline how the use of CIK cells could overcome such drawbacks thanks to their unique features. We highlight the undeniable advantages of using CIK cells as a therapeutic product, underlying the opportunity for further research on the topic.
Collapse
Affiliation(s)
- Elisa Cappuzzello
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Emilia Vigolo
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Giulia D’Accardio
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Department of Hematology, San Bortolo Hospital of Vicenza, Vicenza, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Roberta Sommaggio
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
9
|
Wu CC, Pan MR, Shih SL, Shiau JP, Wu CC, Chang SJ, Kao CN, Chen FM, Hou MF, Luo CW. Combination of FAK inhibitor and cytokine-induced killer cell therapy: An alternative therapeutic strategy for patients with triple-negative breast cancer. Biomed Pharmacother 2023; 163:114732. [PMID: 37254289 DOI: 10.1016/j.biopha.2023.114732] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 06/01/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by the loss of expression of several biomarkers, which limits treatment strategies for the disease. In recent years, immunotherapy has shown promising results in the treatment of various tumors. Emerging evidence demonstrated that TNBC is an immune-activated cancer, suggesting that immunotherapy could be a feasible treatment option for TNBC. Cytokine-induced killer (CIK) cell therapy is considered as a potential treatment for cancer treatment. However, it is still not approved as a standard treatment in the clinical setting. Our previous study demonstrated that focal adhesion kinase (FAK) plays important role in regulating the sensitivity of TNBC cells to CIK cells. In this study, we further verify the role of FAK in regulating the immune response in vivo. Our in vitro study indicated that knockdown of FAK in TNBC cells or treat with the FAK inhibitor followed by co-culture with CIK cells induced more cell death than CIK cells treatment only. RNA-seq analysis indicated that suppression of FAK could affect several immune-related gene expressions in TNBC cells that affects the immune response in the tumor microenvironment of TNBC cells. The combination of FAK inhibitor and CIK cells significantly suppressed tumor growth than the treatment of FAK inhibitor or CIK cells alone in vivo. Our findings provide new insights into the cytotoxic effect of CIK cell therapy in TNBC treatment and indicate that the combination of CIK cell therapy with FAK inhibitors may be an alternative therapeutic strategy for patients with TNBC.
Collapse
Affiliation(s)
- Cheng-Che Wu
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Mei-Ren Pan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Shen-Liang Shih
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Chun-Chieh Wu
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Shu-Jyuan Chang
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Chieh-Ni Kao
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Fang-Ming Chen
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chi-Wen Luo
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Cosmetic Science and Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan.
| |
Collapse
|
10
|
Yu I, Dakwar A, Takabe K. Immunotherapy: Recent Advances and Its Future as a Neoadjuvant, Adjuvant, and Primary Treatment in Colorectal Cancer. Cells 2023; 12:cells12020258. [PMID: 36672193 PMCID: PMC9856401 DOI: 10.3390/cells12020258] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/10/2023] Open
Abstract
Immunotherapy in colorectal cancer (CRC) has made great strides within the past decade. Immune checkpoint inhibitors are a class of immunotherapy and have been shown to greatly improve patient outcomes in mismatch repair-deficient (dMMR) CRC. Now, they are part of the standard of care for this subset of CRC. Because of this, there has been a growing interest in the efficacy and timing of immunotherapy for other subsets of CRC, including locally advanced, metastatic, and microsatellite stable (MSS). In this review, we aim to examine the three main classes of immunotherapy for CRC-immune checkpoint inhibitors (ICIs), adoptive cell transfer therapy (ACT), and tumor vaccines-and discuss the most recent advances and future directions for each.
Collapse
Affiliation(s)
- Irene Yu
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14214, USA
| | - Anthony Dakwar
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14214, USA
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14214, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
- Department of Breast Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
- Correspondence: ; Tel.: +1-716-845-5128
| |
Collapse
|