1
|
You H, Zhao P, Zhao X, Zheng Q, Ma W, Cheng K, Li M, Kou J, Feng W. Promotion of tumor angiogenesis and growth induced by low-dose antineoplastic agents via bone-marrow-derived cells in tumor tissues. Front Pharmacol 2024; 15:1414832. [PMID: 39119610 PMCID: PMC11306047 DOI: 10.3389/fphar.2024.1414832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/17/2024] [Indexed: 08/10/2024] Open
Abstract
Background More research is needed to solidify the basis for reasonable metronomic chemotherapy regimens due to the inconsistent clinical outcomes from studies on metronomic chemotherapy with antineoplastic agents, along with signs of a nonlinear dose-response relationship at low doses. The present study therefore explored the dose-response relationships of representative antineoplastic agents in low dose ranges and their underlying mechanisms. Methods Cyclophosphamide (CPA) and 5-fluorouracil (5-Fu) were employed to observe the effects of the frequent administration of low-dose antineoplastic agents on tumor growth, tumor angiogenesis, and bone-marrow-derived cell (BMDC) mobilization in mouse models. The effects of antineoplastic agents on tumor and endothelial cell functions with or without BMDCs were analyzed in vitro. Results Tumor growth and metastasis were significantly promoted after the administration of CPA or 5-Fu at certain low dose ranges, and were accompanied by enhanced tumor angiogenesis and proangiogenic factor expression in tumor tissues, increased proangiogenic BMDC release in the circulating blood, and augmented proangiogenic BMDC retention in tumor tissues. Low concentrations of CPA or 5-Fu were found to significantly promote tumor cell migration and invasion, and enhance BMDC adhesion to endothelial cells in vitro. Conclusion These results suggest that there are risks in empirical metronomic chemotherapy using low-dose antineoplastic agents and the optimal dosage and administration schedule of antineoplastic agents need to be determined through further research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Weiyi Feng
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
2
|
Shenoy US, Adiga D, Alhedyan F, Kabekkodu SP, Radhakrishnan R. HOXA9 transcription factor is a double-edged sword: from development to cancer progression. Cancer Metastasis Rev 2024; 43:709-728. [PMID: 38062297 PMCID: PMC11156722 DOI: 10.1007/s10555-023-10159-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/30/2023] [Indexed: 04/02/2024]
Abstract
The HOXA9 transcription factor serves as a molecular orchestrator in cancer stemness, epithelial-mesenchymal transition (EMT), metastasis, and generation of the tumor microenvironment in hematological and solid malignancies. However, the multiple modes of regulation, multifaceted functions, and context-dependent interactions responsible for the dual role of HOXA9 as an oncogene or tumor suppressor in cancer remain obscure. Hence, unravelling its molecular complexities, binding partners, and interacting signaling molecules enables us to comprehend HOXA9-mediated transcriptional programs and molecular crosstalk. However, it is imperative to understand its central role in fundamental biological processes such as embryogenesis, foetus implantation, hematopoiesis, endothelial cell proliferation, and tissue homeostasis before designing targeted therapies. Indeed, it presents an enormous challenge for clinicians to selectively target its oncogenic functions or restore tumor-suppressive role without altering normal cellular functions. In addition to its implications in cancer, the present review also focuses on the clinical applications of HOXA9 in recurrence and drug resistance, which may provide a broader understanding beyond oncology, open new avenues for clinicians for accurate diagnoses, and develop personalized treatment strategies. Furthermore, we have also discussed the existing therapeutic options and accompanying challenges in HOXA9-targeted therapies in different cancer types.
Collapse
Affiliation(s)
- U Sangeetha Shenoy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Faisal Alhedyan
- Department of Oral and Maxillofacial Surgery and Diagnostic Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield, United Kingdom
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
3
|
Samy PG, Kanesan J, Badruddin IA, Kamangar S, Ahammad NA. Optimizing chemotherapy treatment outcomes using metaheuristic optimization algorithms: A case study. Biomed Mater Eng 2024; 35:191-204. [PMID: 38143334 DOI: 10.3233/bme-230149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
BACKGROUND This study explores the dynamics of a mathematical model, utilizing ordinary differential equations (ODE), to depict the interplay between cancer cells and effector cells under chemotherapy. The stability of the equilibrium points in the model is analysed using the Jacobian matrix and eigenvalues. Additionally, bifurcation analysis is conducted to determine the optimal values for the control parameters. OBJECTIVE To evaluate the performance of the model and control strategies, benchmarking simulations are performed using the PlatEMO platform. METHODS The Pure Multi-objective Optimal Control Problem (PMOCP) and the Hybrid Multi-objective Optimal Control Problem (HMOCP) are two different forms of optimal control problems that are solved using revolutionary metaheuristic optimisation algorithms. The utilization of the Hypervolume (HV) performance indicator allows for the comparison of various metaheuristic optimization algorithms in their efficacy for solving the PMOCP and HMOCP. RESULTS Results indicate that the MOPSO algorithm excels in solving the HMOCP, with M-MOPSO outperforming for PMOCP in HV analysis. CONCLUSION Despite not directly addressing immediate clinical concerns, these findings indicates that the stability shifts at critical thresholds may impact treatment efficacy.
Collapse
Affiliation(s)
- Prakas Gopal Samy
- Department of Electrical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
- Department of Electrical & Electronics Engineering, Faculty of Engineering, Built Environment & Information Technology, SEGi University & Colleges, Kota Damansara, Petaling Jaya, Selangor, Malaysia
| | - Jeevan Kanesan
- Department of Electrical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Irfan Anjum Badruddin
- Mechanical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
| | - Sarfaraz Kamangar
- Mechanical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
| | - N Ameer Ahammad
- Department of Mathematics, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
4
|
Harrer DC, Lüke F, Pukrop T, Ghibelli L, Reichle A, Heudobler D. Addressing Genetic Tumor Heterogeneity, Post-Therapy Metastatic Spread, Cancer Repopulation, and Development of Acquired Tumor Cell Resistance. Cancers (Basel) 2023; 16:180. [PMID: 38201607 PMCID: PMC10778239 DOI: 10.3390/cancers16010180] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
The concept of post-therapy metastatic spread, cancer repopulation and acquired tumor cell resistance (M-CRAC) rationalizes tumor progression because of tumor cell heterogeneity arising from post-therapy genetic damage and subsequent tissue repair mechanisms. Therapeutic strategies designed to specifically address M-CRAC involve tissue editing approaches, such as low-dose metronomic chemotherapy and the use of transcriptional modulators with or without targeted therapies. Notably, tumor tissue editing holds the potential to treat patients, who are refractory to or relapsing (r/r) after conventional chemotherapy, which is usually based on administering a maximum tolerable dose of a cytostatic drugs. Clinical trials enrolling patients with r/r malignancies, e.g., non-small cell lung cancer, Hodgkin's lymphoma, Langerhans cell histiocytosis and acute myelocytic leukemia, indicate that tissue editing approaches could yield tangible clinical benefit. In contrast to conventional chemotherapy or state-of-the-art precision medicine, tissue editing employs a multi-pronged approach targeting important drivers of M-CRAC across various tumor entities, thereby, simultaneously engaging tumor cell differentiation, immunomodulation, and inflammation control. In this review, we highlight the M-CRAC concept as a major factor in resistance to conventional cancer therapies and discusses tissue editing as a potential treatment.
Collapse
Affiliation(s)
- Dennis Christoph Harrer
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany; (D.C.H.); (F.L.); (T.P.); (D.H.)
| | - Florian Lüke
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany; (D.C.H.); (F.L.); (T.P.); (D.H.)
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Regensburg, Germany
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany; (D.C.H.); (F.L.); (T.P.); (D.H.)
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, 93053 Regensburg, Germany
| | - Lina Ghibelli
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Albrecht Reichle
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany; (D.C.H.); (F.L.); (T.P.); (D.H.)
| | - Daniel Heudobler
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany; (D.C.H.); (F.L.); (T.P.); (D.H.)
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
5
|
Huang X, Ren Q, Yang L, Cui D, Ma C, Zheng Y, Wu J. Immunogenic chemotherapy: great potential for improving response rates. Front Oncol 2023; 13:1308681. [PMID: 38125944 PMCID: PMC10732354 DOI: 10.3389/fonc.2023.1308681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
The activation of anti-tumor immunity is critical in treating cancers. Recent studies indicate that several chemotherapy agents can stimulate anti-tumor immunity by inducing immunogenic cell death and durably eradicate tumors. This suggests that immunogenic chemotherapy holds great potential for improving response rates. However, chemotherapy in practice has only had limited success in inducing long-term survival or cure of cancers when used either alone or in combination with immunotherapy. We think that this is because the importance of dose, schedule, and tumor model dependence of chemotherapy-activated anti-tumor immunity is under-appreciated. Here, we review immune modulation function of representative chemotherapy agents and propose a model of immunogenic chemotherapy-induced long-lasting responses that rely on synergetic interaction between killing tumor cells and inducing anti-tumor immunity. We comb through several chemotherapy treatment schedules, and identify the needs for chemotherapy dose and schedule optimization and combination therapy with immunotherapy when chemotherapy dosage or immune responsiveness is too low. We further review tumor cell intrinsic factors that affect the optimal chemotherapy dose and schedule. Lastly, we review the biomarkers indicating responsiveness to chemotherapy and/or immunotherapy treatments. A deep understanding of how chemotherapy activates anti-tumor immunity and how to monitor its responsiveness can lead to the development of more effective chemotherapy or chemo-immunotherapy, thereby improving the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Xiaojun Huang
- Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qinghuan Ren
- Alberta Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Leixiang Yang
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Center for Reproductive Medicine, Department of Genetic and Genomic Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Di Cui
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chenyang Ma
- Department of Internal Medicine of Traditional Chinese Medicine, The Second People’s Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Yueliang Zheng
- Cancer Center, Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Junjie Wu
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Center for Reproductive Medicine, Department of Genetic and Genomic Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Zou X, Lin X, Cheng H, Chen Y, Wang R, Ma M, Liu Y, Dai Z, Tasiheng Y, Yan Y, Hou Q, Ding F, Chen H, Yu X, Wang X, Liu C. Characterization of intratumoral tertiary lymphoid structures in pancreatic ductal adenocarcinoma: cellular properties and prognostic significance. J Immunother Cancer 2023; 11:e006698. [PMID: 37364934 PMCID: PMC10410893 DOI: 10.1136/jitc-2023-006698] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Tumor-associated tertiary lymphoid structures (TLSs) are functional immune-responsive niches that are not fully understood in pancreatic ductal adenocarcinoma (PDAC). METHODS Fluorescent multiplex immunohistochemistry was performed on sequential sections of surgically resected tumor tissues from 380 PDAC patients without preoperative treatment (surgery alone (SA)) and 136 patients pretreated with neoadjuvant treatment (NAT). Multispectral images were processed via machine learning and image processing platforms, inForm V.2.4 and HALO V.3.2; TLS regions were segmented, and the cells were identified and quantified. The cellular composition and immunological properties of TLSs and their adjacent tissues in PDAC were scored and compared, and their association with prognosis was further examined. RESULTS Intratumoral TLSs were identified in 21.1% (80/380) of patients in the SA group and 15.4% (21/136) of patients in the NAT group. In the SA group, the presence of intratumoral TLSs was significantly associated with improved overall survival (OS) and progression-free survival. The existence of intratumoral TLSs was correlated with elevated levels of infiltrating CD8+T, CD4+T, B cells and activated immune cells in adjacent tissues. A nomogram model was generated with TLS presence as a variable, which successfully predicted PDAC patient OS in an external validation cohort (n=123). In the NAT group, samples exhibited a lower proportion of B cells and a higher proportion of regulatory T cells within intratumoral TLSs. Additionally, these TLSs were smaller in size, with a lower overall maturation level and reduced immune cell activation, and the prognostic value of TLS presence was insignificant in the NAT cohort. CONCLUSION Our study systematically revealed the cellular properties and prognostic values of intratumoral TLSs in PDAC and described the potential impact of NAT on TLS development and function.
Collapse
Affiliation(s)
- Xuan Zou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Xuan Lin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - He Cheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yusheng Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Ruijie Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Mingjian Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yu Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Zhengjie Dai
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yesboli Tasiheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yu Yan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Qinqin Hou
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Fei Ding
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Huan Chen
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Xu Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Radiation Oncology, Cancer Research Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, People's Republic of China
| | - Chen Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
7
|
Masud MA, Kim JY, Kim E. Effective dose window for containing tumor burden under tolerable level. NPJ Syst Biol Appl 2023; 9:17. [PMID: 37221258 DOI: 10.1038/s41540-023-00279-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/05/2023] [Indexed: 05/25/2023] Open
Abstract
A maximum-tolerated dose (MTD) reduces the drug-sensitive cell population, though it may result in the competitive release of drug resistance. Alternative treatment strategies such as adaptive therapy (AT) or dose modulation aim to impose competitive stress on drug-resistant cell populations by maintaining a sufficient number of drug-sensitive cells. However, given the heterogeneous treatment response and tolerable tumor burden level of individual patients, determining an effective dose that can fine-tune competitive stress remains challenging. This study presents a mathematical model-driven approach that determines the plausible existence of an effective dose window (EDW) as a range of doses that conserve sufficient sensitive cells while maintaining the tumor volume below a threshold tolerable tumor volume (TTV). We use a mathematical model that explains intratumor cell competition. Analyzing the model, we derive an EDW determined by TTV and the competitive strength. By applying a fixed endpoint optimal control model, we determine the minimal dose to contain cancer at a TTV. As a proof of concept, we study the existence of EDW for a small cohort of melanoma patients by fitting the model to longitudinal tumor response data. We performed identifiability analysis, and for the patients with uniquely identifiable parameters, we deduced patient-specific EDW and minimal dose. The tumor volume for a patient could be theoretically contained at the TTV either using continuous dose or AT strategy with doses belonging to EDW. Further, we conclude that the lower bound of the EDW approximates the minimum effective dose (MED) for containing tumor volume at the TTV.
Collapse
Affiliation(s)
- M A Masud
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
| | - Jae-Young Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Eunjung Kim
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea.
| |
Collapse
|
8
|
Narendra G, Raju B, Verma H, Kumar M, Jain SK, Tung GK, Thakur S, Kaur R, Kaur S, Sapra B, Singh PK, Silakari O. Raloxifene and bazedoxifene as selective ALDH1A1 inhibitors to ameliorate cyclophosphamide resistance: A drug repurposing approach. Int J Biol Macromol 2023; 242:124749. [PMID: 37160174 DOI: 10.1016/j.ijbiomac.2023.124749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/25/2023] [Accepted: 05/01/2023] [Indexed: 05/11/2023]
Abstract
Cyclophosphamide (CP) is one of the most widely used anticancer drugs for various malignancies. However, its long-term use leads to ALDH1A1-mediated inactivation and subsequent resistance which necessitates the development of potential ALDH1A1 inhibitors. Currently, ALDH1A1 inhibitors from different chemical classes have been reported, but these failed to reach the market due to safety and efficacy problems. Developing a new treatment from the ground requires a huge amount of time, effort, and money, therefore it is worthwhile to improve CP efficacy by proposing better adjuvants as ALDH1A1 inhibitors. Herein, the database constituting the FDA-approved drugs with well-established safety and toxicity profiles was screened through already reported machine learning models by our research group. This model is validated for discriminating the ALDH1A1 inhibitors and non-inhibitors. Virtual screening protocol (VS) from this model identified four FDA-approved drugs, raloxifene, bazedoxifene, avanafil, and betrixaban as selective ALDH1A1 inhibitors. The molecular docking, dynamics, and water swap analysis also suggested these drugs to be promising ALDH1A1 inhibitors which were further validated for their CP resistance reversal potential by in-vitro analysis. The in-vitro enzymatic assay results indicated that raloxifene and bazedoxifene selectively inhibited the ALDH1A1 enzyme with IC50 values of 2.35 and 4.41 μM respectively, whereas IC50 values of both the drugs against ALDH2 and ALDH3A1 was >100 μM. Additional in-vitro stu = dies with well-reported ALDH1A1 overexpressing A549 and MIA paCa-2 cell lines suggested that mafosfamide sensitivity was further ameliorated by the combination of both raloxifene and bazedoxifene. Collectively, in-silico and in-vitro studies indicate raloxifene and bazedoxifene act as promising adjuvants with CP that may improve the quality of treatment for cancer patients with minimal toxicities.
Collapse
Affiliation(s)
- Gera Narendra
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Baddipadige Raju
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Himanshu Verma
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Manoj Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Subheet Kumar Jain
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Gurleen Kaur Tung
- Centre for Basic and Translational Research in Health Sciences, Guru Nanak Dev University, Amritsar, India
| | - Shubham Thakur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Rasdeep Kaur
- Department of Botany and Environmental Sciences, Guru Nanak Dev University, Amritsar, India
| | - Satwinderjeet Kaur
- Department of Botany and Environmental Sciences, Guru Nanak Dev University, Amritsar, India
| | - Bharti Sapra
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Pankaj Kumar Singh
- Integrative Physiology and Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Turku, FI-20520 Turku, Finland
| | - Om Silakari
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India.
| |
Collapse
|
9
|
dePillis L, Caffrey R, Chen G, Dela MD, Eldevik L, McConnell J, Shabahang S, Varvel SA. A mathematical model of the within-host kinetics of SARS-CoV-2 neutralizing antibodies following COVID-19 vaccination. J Theor Biol 2023; 556:111280. [PMID: 36202234 PMCID: PMC9529354 DOI: 10.1016/j.jtbi.2022.111280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 08/22/2022] [Accepted: 09/14/2022] [Indexed: 10/25/2022]
Abstract
Compelling evidence continues to build to support the idea that SARS-CoV-2 Neutralizing Antibody (NAb) levels in an individual can serve as an important indicator of the strength of protective immunity against infection. It is not well understood why NAb levels in some individuals remain high over time, while in others levels decline rapidly. In this work, we present a two-state mathematical model of within-host NAb dynamics in response to vaccination. By fitting only four host-specific parameters, the model is able to capture individual-specific NAb levels over time as measured by the AditxtScore™ for NAbs. The model can serve as a foundation for predicting NAb levels in the long-term, understanding connections between NAb levels, protective immunity, and breakthrough infections, and potentially guiding decisions about whether and when a booster vaccination may be warranted.
Collapse
Affiliation(s)
- Lisette dePillis
- Department of Mathematics, Harvey Mudd College, 301 Platt Blvd., Claremont, CA 91711, United States.
| | - Rebecca Caffrey
- Aditxt, Inc. 737 N. Fifth Street, Suite 200, Richmond, VA 23219, United States
| | - Ge Chen
- Aditxt, Inc. 737 N. Fifth Street, Suite 200, Richmond, VA 23219, United States
| | - Mark D. Dela
- California State Polytechnic University, Pomona, CA, United States
| | - Leif Eldevik
- Aditxt, Inc. 737 N. Fifth Street, Suite 200, Richmond, VA 23219, United States
| | - Joseph McConnell
- Aditxt, Inc. 737 N. Fifth Street, Suite 200, Richmond, VA 23219, United States
| | - Shahrokh Shabahang
- Aditxt, Inc. 737 N. Fifth Street, Suite 200, Richmond, VA 23219, United States
| | - Stephen A. Varvel
- Aditxt, Inc. 737 N. Fifth Street, Suite 200, Richmond, VA 23219, United States
| |
Collapse
|
10
|
Butner JD, Dogra P, Chung C, Pasqualini R, Arap W, Lowengrub J, Cristini V, Wang Z. Mathematical modeling of cancer immunotherapy for personalized clinical translation. NATURE COMPUTATIONAL SCIENCE 2022; 2:785-796. [PMID: 38126024 PMCID: PMC10732566 DOI: 10.1038/s43588-022-00377-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 11/14/2022] [Indexed: 12/23/2023]
Abstract
Encouraging advances are being made in cancer immunotherapy modeling, especially in the key areas of developing personalized treatment strategies based on individual patient parameters, predicting treatment outcomes and optimizing immunotherapy synergy when used in combination with other treatment approaches. Here we present a focused review of the most recent mathematical modeling work on cancer immunotherapy with a focus on clinical translatability. It can be seen that this field is transitioning from pure basic science to applications that can make impactful differences in patients' lives. We discuss how researchers are integrating experimental and clinical data to fully inform models so that they can be applied for clinical predictions, and present the challenges that remain to be overcome if widespread clinical adaptation is to be realized. Lastly, we discuss the most promising future applications and areas that are expected to be the focus of extensive upcoming modeling studies.
Collapse
Affiliation(s)
- Joseph D. Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
- Department of Radiation Oncology, Division of Cancer Biology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - John Lowengrub
- Department of Mathematics, University of California at Irvine, Irvine, CA, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
11
|
Lai V, Neshat SY, Rakoski A, Pitingolo J, Doloff JC. Drug delivery strategies in maximizing anti-angiogenesis and anti-tumor immunity. Adv Drug Deliv Rev 2021; 179:113920. [PMID: 34384826 DOI: 10.1016/j.addr.2021.113920] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/15/2022]
Abstract
Metronomic chemotherapy has been shown to elicit anti-tumor immune response and block tumor angiogenesis distinct from that observed with maximal tolerated dose (MTD) therapy. This review delves into the mechanisms behind anti-tumor immunity and seeks to identify the differential effect of dosing regimens, including daily low-dose and medium-dose intermittent chemotherapy (MEDIC), on both innate and adaptive immune populations involved in observed anti-tumor immune response. Given reports of VEGF/VEGFR blockade antagonizing anti-tumor immunity, drug choice, dose, and selective delivery determined by advanced formulations/vehicles are highlighted as potential sources of innovation for identifying anti-angiogenic modalities that may be combined with metronomic regimens without interrupting key immune players in the anti-tumor response. Engineered drug delivery mechanisms that exhibit extended and local release of anti-angiogenic agents both alone and in combination with chemotherapeutic treatments have also been demonstrated to elicit a potent and potentially systemic anti-tumor immune response, favoring tumor regression and stasis over progression. This review examines this interplay between various cancer models, the host immune response, and select anti-cancer agents depending on drug dosing, scheduling/regimen, and delivery modality.
Collapse
Affiliation(s)
- Victoria Lai
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sarah Y Neshat
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Amanda Rakoski
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - James Pitingolo
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joshua C Doloff
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Materials Science and Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, Division of Cancer Immunology, Sidney Kimmel Comprehensive Cancer Center and the Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
12
|
Muñoz R, Girotti A, Hileeto D, Arias FJ. Metronomic Anti-Cancer Therapy: A Multimodal Therapy Governed by the Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13215414. [PMID: 34771577 PMCID: PMC8582362 DOI: 10.3390/cancers13215414] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Metronomic chemotherapy with different mechanisms of action against cancer cells and their microenvironment represents an exceptional holistic cancer treatment. Each type of tumor has its own characteristics, including each individual tumor in each patient. Understanding the complexity of the dynamic interactions that take place between tumor and stromal cells and the microenvironment in tumor progression and metastases, as well as the response of the host and the tumor itself to anticancer therapy, will allow therapeutic actions with long-lasting effects to be implemented using metronomic regimens. This study aims to highlight the complexity of cellular interactions in the tumor microenvironment and summarize some of the preclinical and clinical results that explain the multimodality of metronomic therapy, which, together with its low toxicity, supports an inhibitory effect on the primary tumor and metastases. We also highlight the possible use of nano-therapeutic agents as good partners for metronomic chemotherapy. Abstract The concept of cancer as a systemic disease, and the therapeutic implications of this, has gained special relevance. This concept encompasses the interactions between tumor and stromal cells and their microenvironment in the complex setting of primary tumors and metastases. These factors determine cellular co-evolution in time and space, contribute to tumor progression, and could counteract therapeutic effects. Additionally, cancer therapies can induce cellular and molecular responses in the tumor and host that allow them to escape therapy and promote tumor progression. In this study, we describe the vascular network, tumor-infiltrated immune cells, and cancer-associated fibroblasts as sources of heterogeneity and plasticity in the tumor microenvironment, and their influence on cancer progression. We also discuss tumor and host responses to the chemotherapy regimen, at the maximum tolerated dose, mainly targeting cancer cells, and a multimodal metronomic chemotherapy approach targeting both cancer cells and their microenvironment. In a combination therapy context, metronomic chemotherapy exhibits antimetastatic efficacy with low toxicity but is not exempt from resistance mechanisms. As such, a better understanding of the interactions between the components of the tumor microenvironment could improve the selection of drug combinations and schedules, as well as the use of nano-therapeutic agents against certain malignancies.
Collapse
Affiliation(s)
- Raquel Muñoz
- Department of Biochemistry, Physiology and Molecular Biology, University of Valladolid, Paseo de Belén, 47011 Valladolid, Spain
- Smart Biodevices for NanoMed Group, University of Valladolid, LUCIA Building, Paseo de Belén, 47011 Valladolid, Spain;
- Correspondence:
| | - Alessandra Girotti
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, LUCIA Building, Paseo de Belén, 47011 Valladolid, Spain;
| | - Denise Hileeto
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON N2L 361, Canada;
| | - Francisco Javier Arias
- Smart Biodevices for NanoMed Group, University of Valladolid, LUCIA Building, Paseo de Belén, 47011 Valladolid, Spain;
| |
Collapse
|
13
|
Frega G, Kepp O, Turchetti D, Rizzo A, Pantaleo MA, Brandi G. Hypothesis on the possible relevance of the immunogenic cell death in the treatment of gestational trophoblastic neoplasms. Transl Oncol 2021; 14:101224. [PMID: 34592590 PMCID: PMC8482038 DOI: 10.1016/j.tranon.2021.101224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/31/2021] [Accepted: 09/10/2021] [Indexed: 11/30/2022] Open
Abstract
The genetic background and the antigenic landscape of cancer cells play a critical role in the response to immunotherapies. A high tumor antigenicity, together with an increased adjuvanticity potentially induced by a peculiar type of cell death, namely immunogenic cell death (ICD), could foster the response to immunogenic therapies. The gestational trophoblastic neoplasm (GTN) is a one-of-a-kind cancer in the oncological landscape due to its exclusive genomic makeup. The prognosis of GTN is significantly better than non-gestational trophoblastic neoplasm (nGTN). Due to its peculiar genetic inheritance, GTN potentially constitutes a singular archetype in the immuno-oncological field.
Collapse
Affiliation(s)
- G Frega
- Department of Specialistic, Diagnostic and Experimental Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Italy; Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni n.15, 40138 Bologna, Italy.
| | - O Kepp
- Equipe labellisée par la Ligue contre le cancer, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - D Turchetti
- Department of Specialistic, Diagnostic and Experimental Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Italy; U.O. Genetica Medica, Policlinico S. Orsola-Malpighi, Centro di Ricerca sui Tumori Ereditari, Bologna Italy
| | - A Rizzo
- Department of Specialistic, Diagnostic and Experimental Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Italy; Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni n.15, 40138 Bologna, Italy
| | - M A Pantaleo
- Department of Specialistic, Diagnostic and Experimental Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Italy; Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni n.15, 40138 Bologna, Italy
| | - G Brandi
- Department of Specialistic, Diagnostic and Experimental Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Italy; Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni n.15, 40138 Bologna, Italy
| |
Collapse
|
14
|
Kareva I, Luddy KA, O’Farrelly C, Gatenby RA, Brown JS. Predator-Prey in Tumor-Immune Interactions: A Wrong Model or Just an Incomplete One? Front Immunol 2021; 12:668221. [PMID: 34531851 PMCID: PMC8438324 DOI: 10.3389/fimmu.2021.668221] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 08/05/2021] [Indexed: 01/05/2023] Open
Abstract
Tumor-immune interactions are often framed as predator-prey. This imperfect analogy describes how immune cells (the predators) hunt and kill immunogenic tumor cells (the prey). It allows for evaluation of tumor cell populations that change over time during immunoediting and it also considers how the immune system changes in response to these alterations. However, two aspects of predator-prey type models are not typically observed in immuno-oncology. The first concerns the conversion of prey killed into predator biomass. In standard predator-prey models, the predator relies on the prey for nutrients, while in the tumor microenvironment the predator and prey compete for resources (e.g. glucose). The second concerns oscillatory dynamics. Standard predator-prey models can show a perpetual cycling in both prey and predator population sizes, while in oncology we see increases in tumor volume and decreases in infiltrating immune cell populations. Here we discuss the applicability of predator-prey models in the context of cancer immunology and evaluate possible causes for discrepancies. Key processes include "safety in numbers", resource availability, time delays, interference competition, and immunoediting. Finally, we propose a way forward to reconcile differences between model predictions and empirical observations. The immune system is not just predator-prey. Like natural food webs, the immune-tumor community of cell types forms an immune-web of different and identifiable interactions.
Collapse
Affiliation(s)
- Irina Kareva
- EMD Serono, Merck KGaA, Billerica, MA, United States
| | - Kimberly A. Luddy
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Cliona O’Farrelly
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Robert A. Gatenby
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Joel S. Brown
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
15
|
Abstract
Choosing and optimizing treatment strategies for cancer requires
capturing its complex dynamics sufficiently well for understanding but
without being overwhelmed. Mathematical models are essential to
achieve this understanding, and we discuss the challenge of choosing
the right level of complexity to address the full range of tumor
complexity from growth, the generation of tumor heterogeneity, and
interactions within tumors and with treatments and the tumor
microenvironment. We discuss the differences between conceptual and
descriptive models, and compare the use of predator-prey models,
evolutionary game theory, and dynamic precision medicine approaches in
the face of uncertainty about mechanisms and parameter values.
Although there is of course no one-size-fits-all approach, we conclude
that broad and flexible thinking about cancer, based on combined
modeling approaches, will play a key role in finding creative and
improved treatments.
Collapse
Affiliation(s)
- Robert A Beckman
- Departments of Oncology and Biostatistics, Bioinformatics, & Biomathematics, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical Informatics, 12231Georgetown University Medical Center, Washington, DC, USA
| | - Irina Kareva
- Mathematical and Computational Sciences Center, School of Human Evolution and Social Change, 7864Arizona State University, Tempe, AZ, USA
| | - Frederick R Adler
- School of Biological Sciences, 415772University of Utah, Salt Lake City, UT, USA.,Department of Mathematics, 415772University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
16
|
Xie Y, Chen X, Li B, Wang X. Comparative efficacy and safety of metronomic chemotherapy in breast cancer: A protocol for network meta-analysis protocol. Medicine (Baltimore) 2021; 100:e26255. [PMID: 34115017 PMCID: PMC8202618 DOI: 10.1097/md.0000000000026255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Metronomic chemotherapy (MC) strategy has been used in breast cancer for more than a decade since it was first proposed. The purpose of this study is to systematically evaluate its efficacy and safety for breast cancer patients at various stages, as well as to clarify the most effective medication strategy when applying MC and discover its most sensitive subpopulation in breast cancer patients. METHOD We will systematically retrieve random controlled trials evaluating the efficacy and safety of MC in breast cancer on PubMed, Cochrane Library, Embase, and web of science to perform this network meta-analysis. Markov chain Monte Carlo method based on Bayesian Theory will be used to conduct network meta-analysis and the efficacy and safety will be ranked by combining direct and indirect evidence in mixed treatment comparisons. We will assess the quality of literatures with the Cochrane Risk Bias Assessment Tool and assess the strength of the evidence using the GRADE methodology. Data analysis will be completed with the WinBUGS, R, Stata and RevMan softwares. RESULTS AND CONCLUSION Through the analysis, we can obtain the ranking of efficacy and safety in different MC strategy, and reveal the specific breast cancer groups that are more sensitive to MC. We access the effectiveness by disease free survival, progress free survival, time to progress, objective response rate, and overall survival, and measure the toxicity by dose-limiting toxicity. The result of our study could provide evidence for clinicians to make a better choice when they consider MC. INPLASY REGISTRATION NUMBER INPLASY202140142.
Collapse
Affiliation(s)
- Ying Xie
- Beijing University of Chinese Medicine, Chaoyang District
| | - Xinjie Chen
- Beijing University of Chinese Medicine, Chaoyang District
| | - Bingxue Li
- Beijing University of Chinese Medicine, Chaoyang District
| | - Xiaoming Wang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Dongcheng District, Beijing, China
| |
Collapse
|
17
|
Effects of Chemotherapy Agents on Circulating Leukocyte Populations: Potential Implications for the Success of CAR-T Cell Therapies. Cancers (Basel) 2021; 13:cancers13092225. [PMID: 34066414 PMCID: PMC8124952 DOI: 10.3390/cancers13092225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/25/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary CAR-T cell therapy is a new approach to cancer treatment that is based on manipulating a patient’s own T cells such that they become able to seek and destroy cancer cells in a highly specific manner. This approach is showing remarkable efficacy in treating some types of blood cancers but so far has been much less effective against solid cancers. Here, we review the diverse effects of chemotherapy agents on circulating leukocyte populations and find that, despite some negative effects over the short term, chemotherapy can favourably modulate the immune systems of cancer patients over the longer term. Since blood is the starting material for CAR-T cell production, we propose that these effects could significantly influence the success of manufacturing, and anti-cancer activity, of CAR-T cells. Thus, if timed correctly, chemotherapy-induced changes to circulating immune cells could allow CAR-T cells to unleash more effective anti-tumour responses. Abstract Adoptive T-cell therapy using autologous T cells genetically modified to express cancer-specific chimeric antigen receptors (CAR) has emerged as a novel approach for cancer treatment. CAR-T cell therapy has been approved in several major jurisdictions for treating refractory or relapsed cases of B-cell precursor acute lymphoblastic leukaemia and diffuse large B-cell lymphoma. However, in solid cancer patients, several clinical studies of CAR-T cell therapy have demonstrated minimal therapeutic effects, thus encouraging interest in better integrating CAR-T cells with other treatments such as conventional cytotoxic chemotherapy. Increasing evidence shows that not only do chemotherapy drugs have tumoricidal effects, but also significantly modulate the immune system. Here, we discuss immunomodulatory effects of chemotherapy drugs on circulating leukocyte populations, including their ability to enhance cytotoxic effects and preserve the frequency of CD8+ T cells and to deplete immunosuppressive populations including regulatory T cells and myeloid-derived suppressor cells. By modulating the abundance and phenotype of leukocytes in the blood (the ‘raw material’ for CAR-T cell manufacturing), we propose that prior chemotherapy could facilitate production of the most effective CAR-T cell products. Further research is required to directly test this concept and identify strategies for the optimal integration of CAR-T cell therapies with cytotoxic chemotherapy for solid cancers.
Collapse
|