1
|
He B, Wood KH, Li ZJ, Ermer JA, Li J, Bastow ER, Sakaram S, Darcy PK, Spalding LJ, Redfern CT, Canes J, Oliveira M, Prat A, Cortes J, Thompson EW, Littlefield BA, Redfern A, Ganss R. Selective tubulin-binding drugs induce pericyte phenotype switching and anti-cancer immunity. EMBO Mol Med 2025; 17:1071-1100. [PMID: 40140727 DOI: 10.1038/s44321-025-00222-6] [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: 10/10/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
The intratumoral immune milieu is crucial for the success of anti-cancer immunotherapy. We show here that stromal modulation by the tubulin-binding anti-cancer drugs combretastatin A4 (CA-4) and eribulin improved tumor perfusion and anti-tumor immunity. This was achieved by reverting highly proliferative, angiogenic pericytes into a quiescent, contractile state which durably normalized the vascular bed and reduced hypoxia in mouse models of pancreatic neuroendocrine cancer, breast cancer and melanoma. The crucial event in pericyte phenotype switching was RhoA kinase activation, which distinguished CA-4 and eribulin effects from other anti-mitotic drugs such as paclitaxel and vinorelbine. Importantly, eribulin pre-treatment sensitized tumors for adoptive T cell therapy or checkpoint inhibition resulting in effector cell infiltration and better survival outcomes in mice. In breast cancer patients, eribulin neoadjuvant treatment induced pericyte maturity and RhoA kinase activity indicating similar vessel remodeling effects as seen in mice. Moreover, a contractile pericyte signature was associated with overall better survival outcome in two independent breast cancer cohorts. This underscores the potential of re-purposing specific anti-cancer drugs to enable synergistic complementation with emerging immunotherapies.
Collapse
Affiliation(s)
- Bo He
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Kira H Wood
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Zhi-Jie Li
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Guangdong, P. R. China
| | - Judith A Ermer
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Ji Li
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Edward R Bastow
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | | | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Lisa J Spalding
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Cameron T Redfern
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Jordi Canes
- SOLTI Cancer Research Group, Barcelona, Spain
| | - Mafalda Oliveira
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Aleix Prat
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Javier Cortes
- SOLTI Cancer Research Group, Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR)-Oncoclínicas&Co, Jersey City, NJ, USA
- Medica Scientia Innovation Research (MEDSIR)-Oncoclínicas&Co, Sao Paulo, Brazil
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
- IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid, Spain
| | - Erik W Thompson
- School of Biomedical Sciences and Centre for Genomics and Personalised Health, Faculty of Health, Queensland University of Technology (QUT) and Translational Research Institute, Brisbane, Australia
| | | | - Andrew Redfern
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- Fiona Stanley Hospital, Perth, WA, Australia
| | - Ruth Ganss
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
2
|
Roncati L. Adjuvant Metronomic Chemotherapy After Surgery in pT1-T2 N0 M0 HER2-Positive and ER/PR-Positive Breast Cancer Plus Targeted Therapy, Anti-Hormonal Therapy, and Radiotherapy, with or Without Immunotherapy: A New Operational Proposal. Cancers (Basel) 2025; 17:1323. [PMID: 40282499 PMCID: PMC12025911 DOI: 10.3390/cancers17081323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Breast cancer is the most common and deadly female-specific malignancy in the world. Four immunohistochemical subtypes are distinguished: luminal A, luminal B, HER2-positive, and triple-negative. In turn, the HER2-positive subtype presents two variants depending on the status of the hormone receptors. The variant that expresses them can benefit from both anti-HER2 and anti-hormonal therapy. Today, MCTP finds application in maintenance therapy after standard of care and in advanced breast cancer when the patient's clinical condition is already seriously compromised by metastatic disease; in this context, it is used as a first-line treatment, in pre-treated subjects, or as a rescue treatment. Here, the use of adjuvant oral MCTP after surgery at an early stage in HER-2 and hormone-positive local breast cancer is proposed, where effective treatment options are available, such as anti-HER2 therapy (e.g., trastuzumab, pertuzumab), anti-hormonal therapy (e.g., tamoxifen, letrozole), radiotherapy, and, in case of strong PD-1 positivity, immunotherapy.
Collapse
Affiliation(s)
- Luca Roncati
- Department of Life Sciences, Health, and Health Care Professions, Link Campus University, 00165 Rome, Italy
| |
Collapse
|
3
|
Gevertz JL, Greene JM, Prosperi S, Comandante-Lou N, Sontag ED. Understanding therapeutic tolerance through a mathematical model of drug-induced resistance. NPJ Syst Biol Appl 2025; 11:30. [PMID: 40204801 PMCID: PMC11982405 DOI: 10.1038/s41540-025-00511-3] [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: 09/03/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
There is growing recognition that phenotypic plasticity enables cancer cells to adapt to various environmental conditions. An example of this adaptability is the ability of an initially sensitive population of cancer cells to acquire resistance and persist in the presence of therapeutic agents. Understanding the implications of this drug-induced resistance is essential for predicting transient and long-term tumor dynamics subject to treatment. This paper introduces a mathematical model of drug-induced resistance which provides excellent fits to time-resolved in vitro experimental data. From observational data of total numbers of cells, the model unravels the relative proportions of sensitive and resistance subpopulations and quantifies their dynamics as a function of drug dose. The predictions are then validated using data on drug doses that were not used when fitting parameters. Optimal control techniques are then utilized to discover dosing strategies that could lead to better outcomes as quantified by lower total cell volume.
Collapse
Affiliation(s)
- Jana L Gevertz
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, USA
| | - James M Greene
- Department of Mathematics, Clarkson University, Potsdam, NY, USA
| | - Samantha Prosperi
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Natacha Comandante-Lou
- Center for Translational & Computational Neuroimmunology, Columbia University Medical Center, New York, NY, USA
| | - Eduardo D Sontag
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, USA.
- Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Gregianin LJ, Rose A, Villarroel M, Almeida MT, Siqueira L, Salgado C, da Costa GA, Castillo L, Santos Pestilho JFC, Brunetto AL. Results of the Latin American Pediatric Oncology Group (GALOP-2011) Trial for Patients With Localized Ewing Sarcoma: A Multicentric Study of Interval-Compressed Multiagent Chemotherapy. Pediatr Blood Cancer 2025; 72:e31554. [PMID: 39838616 DOI: 10.1002/pbc.31554] [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: 09/17/2024] [Revised: 12/17/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND GALOP investigators developed a prospective cooperative protocol for localized Ewing sarcoma (ES) incorporating interval-compressed chemotherapy (VDC/IE, vincristine, doxorubicin, cyclophosphamide/ifosfamide and etoposide). After completing conventional treatment, patients were randomized to 1 year of metronomic chemotherapy (vinblastine and cyclophosphamide). METHODS Phase III randomized prospective trial. Induction consisted of six alternating cycles of VDC/IE every 14 days, followed by local control, and eight cycles of consolidation every 21 days. After consolidation, patients were randomized 1:1 to metronomic chemotherapy or stop treatment, balanced by age (>/< 14 years-old), sex (M/F), site (pelvic/non-pelvic), and size (>/< 8 cm). The results of randomization will be published elsewhere with longer follow-up. RESULTS Between 2011 and 2019, 315 patients (59.7% male, median age 11.0 years) were recruited across 34 centers in Argentina, Brazil, Chile, and Uruguay. The most frequent localizations were axial (45.1%), extremity (38.1%), and pelvic (16.8%). The median time interval between cycles was 19 and 22 days at induction and consolidation, respectively. There were no unexpected toxicity or toxic deaths related to interval compression. The overall response rate post-induction was 81.6%. Local treatment with surgery (50.8%), radiotherapy (19.7%), or a combination (26%) was performed in 304 (96.5%) patients. With a median follow-up of 50 months (range: 1.67-121.7), the 5-year overall and event-free survivals were 68.6% (SE: 0.030) and 63.7% (SE: 0.029), respectively. CONCLUSION Implementation of a multi-institutional protocol with the strategy of interval-compressed induction for ES in South America was feasible with favorable results. This success is attributed to rigorous protocol adherence, extensive educational efforts, and a strong emphasis on data quality maintenance, demonstrating a reproducible model for countries with similar resource limitations.
Collapse
Affiliation(s)
- Lauro José Gregianin
- Service of Pediatric Oncology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Pediatrics, Universidade Federal do Rio Grande do Sul, Brazil
| | - Adriana Rose
- Department of Hematology and Oncology, Hospital de Pediatría Dr J.P. Garrahan, Buenos Aires, Argentina
| | - Milena Villarroel
- Unidad de Oncologia, Hospital Dr. Luis Calvo Mackenna, Providencia, Chile
| | - Maria Tereza Almeida
- Department of Pediatrics, Hospital das Clínicas de São Paulo-ITACI, São Paulo, Brazil
| | - Laurice Siqueira
- Centro de Oncohematologia Pediatrica, Hospital Universitário Oswaldo Cruz, Recife, Brazil
| | - Carmen Salgado
- Department of Pediatric Oncology, Hospital Dr. Exequiel González Cortés, Santigo, Chile
| | | | - Luis Castillo
- Servicio Hemato Oncologico Pediatrico, Centro Hemato-Oncológico Pediátrico, Montevideo, Uruguay
| | | | | |
Collapse
|
5
|
Abdelrady YA, Thabet HS, Sayed AM. The future of metronomic chemotherapy: experimental and computational approaches of drug repurposing. Pharmacol Rep 2025; 77:1-20. [PMID: 39432183 DOI: 10.1007/s43440-024-00662-w] [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: 07/16/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
Metronomic chemotherapy (MC), long-term continuous administration of anticancer drugs, is gaining attention as an alternative to the traditional maximum tolerated dose (MTD) chemotherapy. By combining MC with other treatments, the therapeutic efficacy is enhanced while minimizing toxicity. MC employs multiple mechanisms, making it a versatile approach against various cancers. However, drug resistance limits the long-term effectiveness of MC, necessitating ongoing development of anticancer drugs. Traditional drug discovery is lengthy and costly due to processes like target protein identification, virtual screening, lead optimization, and safety and efficacy evaluations. Drug repurposing (DR), which screens FDA-approved drugs for new uses, is emerging as a cost-effective alternative. Both experimental and computational methods, such as protein binding assays, in vitro cytotoxicity tests, structure-based screening, and several types of association analyses (Similarity-Based, Network-Based, and Target Gene), along with retrospective clinical analyses, are employed for virtual screening. This review covers the mechanisms of MC, its application in various cancers, DR strategies, examples of repurposed drugs, and the associated challenges and future directions.
Collapse
Affiliation(s)
- Yousef A Abdelrady
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104, Freiburg, Germany
| | - Hayam S Thabet
- Microbiology Department, Faculty of Veterinary Medicine, Assiut University, Asyut, 71526, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Asyut, 71516, Egypt
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Kingdom of Saudi Arabia
| |
Collapse
|
6
|
Madabhavi IV, Sarkar MS, Sagar RD. Metronomic Chemotherapy After Palliative Radiotherapy in Rare Soft Tissue Sarcomas: An Insight into Radiation Therapy and the Immune Response. Indian J Surg Oncol 2024; 15:677-683. [PMID: 39555377 PMCID: PMC11564421 DOI: 10.1007/s13193-024-01966-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 05/19/2024] [Indexed: 11/19/2024] Open
Abstract
Soft tissue sarcomas (STSs) account for less than 1% of the overall human burden of malignant tumors. The intent of treatment in metastatic STSs is palliative and prognosis is dismal. This study aims to evaluate the role of low-dose radiotherapy and low-dose oral metronomic therapy in heavily pretreated metastatic infrequent STSs. This study was conducted in a prospective observational manner in a tertiary care center. A total of 16 cases met the inclusion criteria for enrollment in the study group. The diagnosis of all subtypes of STS was confirmed by histopathology and immunohistochemistry or cytogenetic studies. All the enrolled patients with metastatic STSs who were treated with surgery and two lines of chemotherapy were initially treated with low-dose radiotherapy of 20 Gy in 5 fractions or 30 Gy in 10 fractions according to the ECOG performance status of the patient. Out of 16 patients, seven patients (43.75%) were treated with 20 Gy, 5#, and 9 patients (56.25%) were treated with 30 Gy, 10# radiotherapy (RT). Out of 16 patients, four were of malignant fibrohistiocytic sarcoma, three were angiosarcoma, three were malignant phylloides tumor of the breast, two were fibrosarcoma, and one patient from each of the following subtypes inflammatory myofiroblastic tumor, leiomyosarcoma, synovial sarcoma, and dermato fibrosarcoma protuberance. Out of 16 patients, seven (43.75%) had a partial response (PR), nine (56.25%) had stable disease (SD) at 3 months, and all the patients had SD at 6 months of evaluation. All the patients had enjoyed a better quality of life as compared to their life during injectable chemotherapy. Targeted low-dose radiation and metronomic chemotherapy leads to quantifiable antiangiogeneic effects and immune effects in the local tumor microenvironment, and influences circulating immune mediators and anti-angiogenesis that could potentially help eradicate disease both within and outside the radiation treatment field.
Collapse
Affiliation(s)
- Irappa V. Madabhavi
- Department of Medical and Pediatric Oncology, J N Medical College, and KLE Academy of Higher Education and Research (KAHER), Belagavi, Karnataka India
- Kerudi Cancer Hospital, Bagalkot, Karnataka India
- Nanjappa Hospital, Davanagere, Karnataka India
- Dr. N B Patil Hospital, Gadag, Karnataka India
| | - Malay S. Sarkar
- Department of Pulmonary Medicine, Indira Gandhi Medical College, Shimla, Himachal Pradesh India
| | - Raghavendra D. Sagar
- Department of Radiation Oncology, J N Medical College, Belagavi, Karnataka India
| |
Collapse
|
7
|
Sahoo GP, Rai VK, Pradhan D, Halder J, Rajwar TK, Mahanty R, Saha I, Mishra A, Dash P, Dash C, Al-Tamimi J, Manoharadas S, Kar B, Ghosh G, Rath G. A doxorubicin loaded chitosan-poloxamer in situ implant for the treatment of breast cancer. RSC Adv 2024; 14:33952-33967. [PMID: 39463476 PMCID: PMC11503159 DOI: 10.1039/d4ra06253a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024] Open
Abstract
Breast cancer is a serious concern for many women worldwide. Drug-loaded implants have shown several benefits over systemic administrations. To provide anti-cancer drugs with controlled release and reduced systemic toxicity, biodegradable in situ implants have attracted a lot of attention. In the present study, we aimed to design and optimize a doxorubicin-loaded chitosan-poloxamer in situ implant for breast cancer treatment. Utilizing Box-Behnken Design and a Quality-by-Design (QbD) methodology, the in situ implant was prepared with chitosan (X1), poloxamer 407 concentration (X2), and stirring time (X3) as the independent variables. It was characterized for its in vitro gelation time, pH, rheology, and morphology, and evaluated based on drug release profile, in vitro cytotoxicity activities, in vitro anti-inflammatory potential, in vitro cellular uptake, and in vivo anti-inflammatory and pharmacokinetics to ensure their therapeutic outcomes. The results revealed that the prepared formulation showed a gelation time of 26 ± 0.2 s with a viscosity of 8312.6 ± 114.2 cPs at 37 °C. The developed formulation showed better cytotoxic activity in MCF-7 cell lines compared to the free drug solution. It demonstrated reduced levels of pro-inflammatory cytokines in RAW 264.7 macrophages. Further, the prepared in situ implant increases the intracellular accumulation of DOX in the MCF-7 cells. The in vivo pharmacokinetic investigations depicted an increase in t 1/2 and a decrease in AUC of the developed formulation resulting in prolonged drug release and there could be a lower drug concentration in the bloodstream than for the free drug. Therefore, the developed in situ implant may offer a viable option for breast cancer treatment.
Collapse
Affiliation(s)
- Guru Prasanna Sahoo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Vineet Kumar Rai
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Deepak Pradhan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Jitu Halder
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Tushar Kanti Rajwar
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Ritu Mahanty
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Ivy Saha
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Ajit Mishra
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Priyanka Dash
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Chandan Dash
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Jameel Al-Tamimi
- Zoology Department, College of Science, King Saud University P. O. Box 2455 Riyadh Saudi Arabia
| | - Salim Manoharadas
- Department of Botany and Microbiology, College of Science, King Saud University P. O. Box. 2454 Riyadh 11451 Saudi Arabia
| | - Biswakanth Kar
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Goutam Ghosh
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Goutam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| |
Collapse
|
8
|
Garlisi B, Lauks S, Aitken C, Ogilvie LM, Lockington C, Petrik D, Eichhorn JS, Petrik J. The Complex Tumor Microenvironment in Ovarian Cancer: Therapeutic Challenges and Opportunities. Curr Oncol 2024; 31:3826-3844. [PMID: 39057155 PMCID: PMC11275383 DOI: 10.3390/curroncol31070283] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
The tumor microenvironment (TME) in ovarian cancer (OC) has much greater complexity than previously understood. In response to aggressive pro-angiogenic stimulus, blood vessels form rapidly and are dysfunctional, resulting in poor perfusion, tissue hypoxia, and leakiness, which leads to increased interstitial fluid pressure (IFP). Decreased perfusion and high IFP significantly inhibit the uptake of therapies into the tumor. Within the TME, there are numerous inhibitor cells, such as myeloid-derived suppressor cells (MDSCs), tumor association macrophages (TAMs), regulatory T cells (Tregs), and cancer-associated fibroblasts (CAFs) that secrete high numbers of immunosuppressive cytokines. This immunosuppressive environment is thought to contribute to the lack of success of immunotherapies such as immune checkpoint inhibitor (ICI) treatment. This review discusses the components of the TME in OC, how these characteristics impede therapeutic efficacy, and some strategies to alleviate this inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (B.G.); (S.L.); (C.A.); (L.M.O.); (C.L.); (D.P.); (J.S.E.)
| |
Collapse
|
9
|
Petrucci GN, Magalhães TR, Dias M, Queiroga FL. Metronomic chemotherapy: bridging theory to clinical application in canine and feline oncology. Front Vet Sci 2024; 11:1397376. [PMID: 38903691 PMCID: PMC11187343 DOI: 10.3389/fvets.2024.1397376] [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: 03/07/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024] Open
Abstract
Veterinary oncology has experienced significant evolution over the last few decades, with chemotherapy being currently applied to several neoplasms with therapeutic success. Traditionally, chemotherapy protocols are based on classic cytostatic drugs under the concept of maximum tolerated dose (MTD), which has been associated with a greater risk of toxicity and resistance. Thus, new therapeutic alternatives have emerged, such as metronomic chemotherapy (MC), introducing a new paradigm in cancer treatment. MC consists of administering low doses of chemotherapy drugs continuously over a long period of time, modulating the tumour microenvironment (TME) due to the combination of cytotoxic, antiangiogenic and immunomodulatory effects. This multi-targeted therapy has been described as a treatment option in several canine and feline cancers since 2007, with positive results already published in the literature, particularly in mammary carcinomas and soft tissue sarcomas in dogs. The aim of this review article is to describe the current knowledge about the use of MC in small animal oncology, with emphasis on its mechanisms of action, the most commonly used drugs and clinical outcome.
Collapse
Affiliation(s)
- Gonçalo N. Petrucci
- Onevet Hospital Veterinário do Porto, Porto, Portugal
- Animal and Veterinary Department, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
- Animal and Veterinary Research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Department of Veterinary Sciences, Center for Investigation Vasco da Gama (CIVG), Vasco da Gama University School (EUVG), Coimbra, Portugal
| | - Tomás Rodrigues Magalhães
- Animal and Veterinary Research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Márcia Dias
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Felisbina Luísa Queiroga
- Animal and Veterinary Research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Centre for the Study of Animal Science, CECA-ICETA, University of Porto, Porto, Portugal
| |
Collapse
|
10
|
Basar OY, Mohammed S, Qoronfleh MW, Acar A. Optimizing cancer therapy: a review of the multifaceted effects of metronomic chemotherapy. Front Cell Dev Biol 2024; 12:1369597. [PMID: 38813084 PMCID: PMC11133583 DOI: 10.3389/fcell.2024.1369597] [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: 01/12/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
Metronomic chemotherapy (MCT), characterized by the continuous administration of chemotherapeutics at a lower dose without prolonged drug-free periods, has garnered significant attention over the last 2 decades. Extensive evidence from both pre-clinical and clinical settings indicates that MCT induces distinct biological effects than the standard Maximum Tolerated Dose (MTD) chemotherapy. The low toxicity profile, reduced likelihood of inducing acquired therapeutic resistance, and low cost of MCT render it an attractive chemotherapeutic regimen option. One of the most prominent aspects of MCT is its anti-angiogenesis effects. It has been shown to stimulate the expression of anti-angiogenic molecules, thereby inhibiting angiogenesis. In addition, MCT has been shown to decrease the regulatory T-cell population and promote anti-tumor immune response through inducing dendritic cell maturation and increasing the number of cytotoxic T-cells. Combination therapies utilizing MCT along with oncolytic virotherapy, radiotherapy or other chemotherapeutic regimens have been studied extensively. This review provides an overview of the current status of MCT research and the established mechanisms of action of MCT treatment and also offers insights into potential avenues of development for MCT in the future.
Collapse
Affiliation(s)
- Oyku Yagmur Basar
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Sawsan Mohammed
- Qatar University, QU Health, College of Medicine, Doha, Qatar
| | - M. Walid Qoronfleh
- Q3 Research Institute (QRI), Research and Policy Division, Ypsilanti, MI, United States
| | - Ahmet Acar
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| |
Collapse
|
11
|
Mayer EL, Tayob N, Ren S, Savoie JJ, Spigel DR, Burris HA, Ryan PD, Harris LN, Winer EP, Burstein HJ. A randomized phase II study of metronomic cyclophosphamide and methotrexate (CM) with or without bevacizumab in patients with advanced breast cancer. Breast Cancer Res Treat 2024; 204:123-132. [PMID: 38019444 DOI: 10.1007/s10549-023-07167-9] [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: 08/22/2023] [Accepted: 10/24/2023] [Indexed: 11/30/2023]
Abstract
PURPOSE Metronomic chemotherapy has the potential to offer tumor control with reduced toxicity when compared to standard dose chemotherapy in patients with metastatic breast cancer. As metronomic chemotherapy may target the tumor microvasculature, it has the potential for synergistic effects with antiangiogenic agents such as the VEGF-A inhibitor bevacizumab. METHODS In this randomized phase II study, patients with metastatic breast cancer were randomized to receive metronomic oral cyclophosphamide and methotrexate (CM) combined with bevacizumab (Arm A) or CM alone (Arm B). The primary endpoint was objective response rate (ORR). Secondary endpoints included progression-free survival (PFS), overall survival (OS), and safety and tolerability. RESULTS A total of 55 patients were enrolled, with 34 patients treated on Arm A and 21 patients treated on Arm B. The ORR was modestly higher in Arm A (26%) than in Arm B (10%); neither met the 40% cutoff for further clinical evaluation. The median time to progression (TTP) was 5.52 months (3.22-13.6) on Arm A and 1.82 months (1.54-6.70) on Arm B (log-rank p = 0.008). The median OS was 29.6 months (17.2-NA) on Arm A and 16.2 months (15.7-NA) on Arm B (log-rank p = 0.7). Common all-grade adverse events in both arms included nausea, fatigue, and elevated AST. CONCLUSION The combination of metronomic CM with bevacizumab significantly improved PFS over CM alone, although there was no significant difference in OS. Oral metronomic chemotherapy alone has limited activity in advanced breast cancer. CLINICALTRIALS gov Identifier: NCT00083031. Date of Registration: May 17, 2004.
Collapse
Affiliation(s)
- Erica L Mayer
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Nabihah Tayob
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Siyang Ren
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Jennifer J Savoie
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - David R Spigel
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Sarah Cannon Research Institute, Nashville, TN, USA
| | - Howard A Burris
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Sarah Cannon Research Institute, Nashville, TN, USA
| | - Paula D Ryan
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Texas Oncology, The Woodlands, TX, USA
| | - Lyndsay N Harris
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- National Cancer Institute, Bethesda, MD, USA
| | - Eric P Winer
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Yale Cancer Center, New Haven, CT, USA
| | - Harold J Burstein
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA.
| |
Collapse
|
12
|
Moi D, Bonanni D, Belluti S, Linciano P, Citarella A, Franchini S, Sorbi C, Imbriano C, Pinzi L, Rastelli G. Discovery of potent pyrrolo-pyrimidine and purine HDAC inhibitors for the treatment of advanced prostate cancer. Eur J Med Chem 2023; 260:115730. [PMID: 37633202 DOI: 10.1016/j.ejmech.2023.115730] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/01/2023] [Accepted: 08/14/2023] [Indexed: 08/28/2023]
Abstract
The development of drugs for the treatment of advanced prostate cancer (PCA) remains a challenging task. In this study we have designed, synthesized and tested twenty-nine novel HDAC inhibitors based on three different zinc binding groups (trifluoromethyloxadiazole, hydroxamic acid, and 2-mercaptoacetamide). These warheads were conveniently tethered to variously substituted phenyl linkers and decorated with differently substituted pyrrolo-pyrimidine and purine cap groups. Remarkably, most of the compounds showed nanomolar inhibitory activity against HDAC6. To provide structural insights into the Structure-Activity Relationships (SAR) of the investigated compounds, docking of representative inhibitors and molecular dynamics of HDAC6-inhibitor complexes were performed. Compounds of the trifluoromethyloxadiazole and hydroxamic acid series exhibited promising anti-proliferative activities, HDAC6 targeting in PCA cells, and in vitro tumor selectivity. Representative compounds of the two series were tested for solubility, cell permeability and metabolic stability, demonstrating favorable in vitro drug-like properties. The more interesting compounds were subjected to migration assays, which revealed that compound 13 and, to a lesser extent, compound 15 inhibited the invasive behaviour of androgen-sensitive and -insensitive advanced prostate cancer cells. Compound 13 was profiled against all HDACs and found to inhibit all members of class II HDACs (except for HDAC10) and to be selective with respect to class I and class IV HDACs. Overall, compound 13 combines potent inhibitory activity and class II selectivity with favorable drug-like properties, an excellent anti-proliferative activity and marked anti-migration properties on PCA cells, making it an excellent lead candidate for further optimization.
Collapse
Affiliation(s)
- Davide Moi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Davide Bonanni
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Pasquale Linciano
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Andrea Citarella
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Silvia Franchini
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Claudia Sorbi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy.
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy.
| |
Collapse
|
13
|
Muraro E, Vinante L, Fratta E, Bearz A, Höfler D, Steffan A, Baboci L. Metronomic Chemotherapy: Anti-Tumor Pathways and Combination with Immune Checkpoint Inhibitors. Cancers (Basel) 2023; 15:2471. [PMID: 37173937 PMCID: PMC10177461 DOI: 10.3390/cancers15092471] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Increasing evidence pinpoints metronomic chemotherapy, a frequent and low dose drug administration with no prolonged drug-free intervals, as a potential tool to fight certain types of cancers. The primary identified targets of metronomic chemotherapy were the tumor endothelial cells involved in angiogenesis. After this, metronomic chemotherapy has been shown to efficiently target the heterogeneous population of tumor cells and, more importantly, elicit the innate and adaptive immune system reverting the "cold" to "hot" tumor immunologic phenotype. Although metronomic chemotherapy is primarily used in the context of a palliative setting, with the development of new immunotherapeutic drugs, a synergistic therapeutic role of the combined metronomic chemotherapy and immune checkpoint inhibitors has emerged at both the preclinical and clinical levels. However, some aspects, such as the dose and the most effective scheduling, still remain unknown and need further investigation. Here, we summarize what is currently known of the underlying anti-tumor effects of the metronomic chemotherapy, the importance of the optimal therapeutic dose and time-exposure, and the potential therapeutic effect of the combined administration of metronomic chemotherapy with checkpoint inhibitors in preclinical and clinical settings.
Collapse
Affiliation(s)
- Elena Muraro
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.M.); (E.F.); (A.S.)
| | - Lorenzo Vinante
- Radiation Oncology Department, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy;
| | - Elisabetta Fratta
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.M.); (E.F.); (A.S.)
| | - Alessandra Bearz
- Medical Oncology Department, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy;
| | - Daniela Höfler
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.M.); (E.F.); (A.S.)
| | - Lorena Baboci
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.M.); (E.F.); (A.S.)
| |
Collapse
|
14
|
Panthi VK, Dua K, Singh SK, Gupta G, Hansbro PM, Paudel KR. Nanoformulations-Based Metronomic Chemotherapy: Mechanism, Challenges, Recent Advances, and Future Perspectives. Pharmaceutics 2023; 15:pharmaceutics15041192. [PMID: 37111677 PMCID: PMC10146318 DOI: 10.3390/pharmaceutics15041192] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Cancer-related death is a significant health and economic burden worldwide, and some conventional chemotherapy is associated with limited effectiveness in completely curing various cancers, severe adverse effects, and destruction of healthy cells. To overcome the complications associated with conventional treatment, metronomic chemotherapy (MCT) is extensively suggested. In this review, we aim to highlight the importance of MCT over conventional chemotherapeutic approach with emphasis on nanoformulations-based MCT, their mechanism, challenges, recent advances, and future perspectives. Nanoformulations-based MCT revealed remarkable antitumor activity in both preclinical and clinical settings. For example, the metronomic scheduling of oxaliplatin-loaded nanoemulsion and polyethylene glycol-coated stealth nanoparticles incorporating paclitaxel were proven very effective in tumor-bearing mice and rats, respectively. Additionally, several clinical studies have demonstrated the benefit of MCT with acceptable tolerance. Moreover, metronomic might be a promising treatment strategy for improving cancer care in low- and middle-income nations. However, an appropriate alternative to a metronomic regimen for an individual ailment, suitable combinational delivery and scheduling, and predictive biomarkers are certain parts that remain unanswered. Further clinical-based comparative research studies are mandatory to be performed before entailing this treatment modality in clinical practice as alternative maintenance therapy or in place of transferring to therapeutic management.
Collapse
Affiliation(s)
- Vijay Kumar Panthi
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur 302017, India
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia
| |
Collapse
|
15
|
Metronomic Chemotherapy Based on Topotecan or Topotecan and Cyclophosphamide Combination (CyTo) in Advanced, Pretreated Ovarian Cancer. Cancers (Basel) 2023; 15:cancers15041067. [PMID: 36831410 PMCID: PMC9954570 DOI: 10.3390/cancers15041067] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Patients with advanced ovarian cancer (OC) have a detrimental prognosis. The options for systemic treatment of advanced OC in later lines of treatment are limited by the availability of active therapies and their applicability to often fragile, exhausted patients with poor performance status. Metronomic chemotherapy (MC) is a concept of a continuous administration of cytotoxic drugs, which is characterized by multidirectional activity (anti-proliferative, anti-angiogenic, and anti-immunosuppressive) and low toxicity. We have performed a retrospective analysis of consecutive, advanced, chemo-refractory OC patients treated with MC based on single-agent topotecan (1 mg p.o. q2d) or on a topotecan (1 mg q2d) and cyclophosphamide (50 mg p.o. qd) combination (CyTo). Metronomic chemotherapy demonstrated promising activity, with 72% and 86% of patients achieving biochemical or objective disease control and 18% and 27% of patients achieving a biochemical or objective response, respectively. The median PFS in the whole population was 3.65 months, but the median PFS in patients with a biochemical response to MC (18.2% of patients) reached 10.7 months. The study also suggested that overweight or obese patients had significantly better outcomes on MC than patients with BMI <25 kg/m2. This article is the first report in the literature on metronomic chemotherapy based on a topotecan + cyclophosphamide combination (CyTo). The CyTo regimen demonstrated safety, clinical activity, and potential broad clinical applicability in advanced OC patients and will be evaluated in a forthcoming clinical trial.
Collapse
|
16
|
Kamal MV, Rao M, Damerla RR, Pai A, Sharan K, Palod A, Shetty PS, Usman N, Kumar NAN. A Mechanistic Review of Methotrexate and Celecoxib as a Potential Metronomic Chemotherapy for Oral Squamous Cell Carcinoma. Cancer Invest 2023; 41:144-154. [PMID: 36269850 DOI: 10.1080/07357907.2022.2139840] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The combination of low-dose methotrexate and celecoxib as metronomic chemotherapy (MCT) is a novel therapy, believed to act by modulating the immune response, inhibiting angiogenesis and its cytotoxic action, though the exact mechanism of action is unclear. Clinically, MCT was found to be very effective in delaying tumor progression in patients with head and neck squamous cell carcinoma in both curative and palliative settings. This review was aimed to give a brief insight into the mechanism of action and potential molecular alterations of MCT in the treatment of oral cancers taking into consideration the various in vivo and in vitro studies.
Collapse
Affiliation(s)
- Mehta Vedant Kamal
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Rama Rao Damerla
- Department of Medical Genetics, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Ananth Pai
- Department of Medical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Krishan Sharan
- Department of Radiotherapy and Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Akhil Palod
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Preethi S Shetty
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Nawaz Usman
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Naveena A N Kumar
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| |
Collapse
|
17
|
Alshammari MK, Alghazwni MK, Alharbi AS, Alqurashi GG, Kamal M, Alnufaie SR, Alshammari SS, Alshehri BA, Tayeb RH, Bougeis RJM, Aljehani AA, Alotaibi NM, Abida A, Imran M. Nanoplatform for the Delivery of Topotecan in the Cancer Milieu: An Appraisal of its Therapeutic Efficacy. Cancers (Basel) 2022; 15:cancers15010065. [PMID: 36612067 PMCID: PMC9817931 DOI: 10.3390/cancers15010065] [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: 11/25/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Chemotherapy has been the predominant treatment modality for cancer patients, but its overall performance is still modest. Difficulty in penetration of tumor tissues, a toxic profile in high doses, multidrug resistance in an array of tumor types, and the differential architecture of tumor cells as they grow are some of the bottlenecks associated with the clinical usage of chemotherapeutics. Recent advances in tumor biology understanding and the emergence of novel targeted drug delivery tools leveraging various nanosystems offer hope for developing effective cancer treatments. Topotecan is a topoisomerase I inhibitor that stabilizes the transient TOPO I-DNA cleavable complex, leading to single-stranded breaks in DNA. Due to its novel mechanism of action, TOPO is reported to be active against various carcinomas, namely small cell lung cancer, cervical cancer, breast cancer, and ovarian cancer. Issues of cross-resistance with numerous drugs, rapid conversion to its inactive form in biological systems, appended adverse effects, and higher water solubility limit its therapeutic efficacy in clinical settings. Topotecan nanoformulations offer several benefits for enhancing the therapeutic action of this significant class of chemotherapeutics. The likelihood that the target cancer cells will be exposed to the chemotherapeutic drug while in the drug-sensitive s-phase is increased due to the slow and sustained release of the chemotherapeutic, which could provide for a sustained duration of exposure of the target cancer cells to the bioavailable drug and result in the desired therapeutic outcome. This article explores nanoenabled active and passive targeting strategies and combinatorial therapy employing topotecan to ameliorate various cancers, along with a glimpse of the clinical studies utilizing the said molecule.
Collapse
Affiliation(s)
- Mohammed Kanan Alshammari
- Department of Clinical Pharmacy, King Fahad Medical City, Riyadh 12211, Saudi Arabia
- Correspondence: (M.K.A.); (M.I.)
| | | | - Abrar Saleh Alharbi
- Department of Pharmaceutical Sciences, Maternity and Children’s Hospital, Mecca 24246, Saudi Arabia
| | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Salman Rahim Alnufaie
- Department of Infection Control, Riyadh Third Health Cluster, Riyadh 13223, Saudi Arabia
| | - Salem Sayer Alshammari
- Department of Pharmaceutical Care, Al-Dawaa Medical Services, Jubail 35412, Saudi Arabia
| | - Bandar Ali Alshehri
- Laboratory Department, King Abdulaziz University Hospital, Jeddah 21589, Saudi Arabia
| | - Rami Hatem Tayeb
- Laboratory Department, King Abdulaziz University Hospital, Jeddah 21589, Saudi Arabia
| | | | - Alaa Adel Aljehani
- Laboratory Department, King Abdulaziz University Hospital, Jeddah 21589, Saudi Arabia
| | - Nawaf M. Alotaibi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Abida Abida
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mohd. Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
- Correspondence: (M.K.A.); (M.I.)
| |
Collapse
|
18
|
Programmed Cell Death-Ligand 1 in Head and Neck Squamous Cell Carcinoma: Molecular Insights, Preclinical and Clinical Data, and Therapies. Int J Mol Sci 2022; 23:ijms232315384. [PMID: 36499710 PMCID: PMC9738355 DOI: 10.3390/ijms232315384] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/24/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Aberrant expression of the programmed cell death protein ligand 1 (PD-L1) constitutes one of the main immune evasion mechanisms of cancer cells. The approval of drugs against the PD-1-PD-L1 axis has given new impetus to the chemo-therapy of many malignancies. We performed a literature review from 1992 to August 2022, summarizing evidence regarding molecular structures, physiological and pathological roles, mechanisms of PD-L1 overexpression, and immunotherapy evasion. Furthermore, we summarized the studies concerning head and neck squamous cell carcinomas (HNSCC) immunotherapy and the prospects for improving the associated outcomes, such as identifying treatment response biomarkers, new pharmacological combinations, and new molecules. PD-L1 overexpression can occur via four mechanisms: genetic modifications; inflammatory signaling; oncogenic pathways; microRNA or protein-level regulation. Four molecular mechanisms of resistance to immunotherapy have been identified: tumor cell adaptation; changes in T-cell function or proliferation; alterations of the tumor microenvironment; alternative immunological checkpoints. Immunotherapy was indeed shown to be superior to traditional chemotherapy in locally advanced/recurrent/metastatic HNSCC treatments.
Collapse
|
19
|
Shet D, Gatty RC. Impressive response to oral metronomic chemotherapy in ovarian cancer. CURRENT PROBLEMS IN CANCER: CASE REPORTS 2022. [DOI: 10.1016/j.cpccr.2022.100197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
20
|
Scagliotti A, Capizzi L, Cazzaniga ME, Ilari A, De Giorgi M, Cordani N, Gallazzi M, Bruno A, Pelosi G, Albini A, Lavitrano M, Grassilli E, Cerrito MG. Co-targeting triple-negative breast cancer cells and endothelial cells by metronomic chemotherapy inhibits cell regrowth and migration via downregulation of the FAK/VEGFR2/VEGF axis and autophagy/apoptosis activation. Front Oncol 2022; 12:998274. [PMID: 36531071 PMCID: PMC9749857 DOI: 10.3389/fonc.2022.998274] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/14/2022] [Indexed: 12/07/2023] Open
Abstract
High-dose standard-of-care chemotherapy is the only option for triple-negative breast cancer (TNBC) patients, which eventually die due to metastatic tumors. Recently, metronomic chemotherapy (mCHT) showed advantages in treating TNBCs leading us to investigate the anti-metastatic and anti-angiogenic potential of metronomic 5-Fluorouracil plus Vinorelbine (5-FU+VNR) on endothelial cells (ECs) and TNBCs in comparison to standard treatment (STD). We found that 10-fold lower doses of 5-FU+VNR given mCHT vs. STD inhibits cell proliferation and survival of ECs and TNBC cells. Both schedules strongly affect ECs migration and invasion, but in TNBC cells mCHT is significantly more effective than STD in impairing cell migration and invasion. The two treatments disrupt FAK/VEGFR/VEGF signaling in both ECs and TNBC cells. mCHT, and to a much lesser extent STD treatment, induces apoptosis in ECs, whereas it switches the route of cell death from apoptosis (as induced by STD) to autophagy in TNBC cells. mCHT-treated TNBCs-derived conditioned medium also strongly affects ECs' migration, modulates different angiogenesis-associated proteins, and hampers angiogenesis in matrix sponge in vivo. In conclusion, mCHT administration of 5-FU+VNR is more effective than STD schedule in controlling cell proliferation/survival and migration/invasion of both ECs and TNBC cells and has a strong anti-angiogenic effect. Our data suggest that the stabilization of tumor growth observed in TNBC patients treated with mCHT therapy schedule is likely due not only to direct cytotoxic effects but also to anti-metastatic and anti-angiogenic effects.
Collapse
Affiliation(s)
- Arianna Scagliotti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Laura Capizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Marina Elena Cazzaniga
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Phase 1 Research Center, Azienda Socio Sanitaria Territoriale (ASST) di Monza, Monza, Italy
| | - Alice Ilari
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Marco De Giorgi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Nicoletta Cordani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Matteo Gallazzi
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Antonino Bruno
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Giuseppe Pelosi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Adriana Albini
- IRCCS European Institute of Oncology (IEO), Milan, Italy
| | | | - Emanuela Grassilli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | |
Collapse
|
21
|
Deb D, Zhu S, LeBlanc MJ, Danino T. Assessing chemotherapy dosing strategies in a spatial cell culture model. Front Oncol 2022; 12:980770. [PMID: 36505801 PMCID: PMC9729937 DOI: 10.3389/fonc.2022.980770] [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: 06/28/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Predicting patient responses to chemotherapy regimens is a major challenge in cancer treatment. Experimental model systems coupled with quantitative mathematical models to calculate optimal dose and frequency of drugs can enable improved chemotherapy regimens. Here we developed a simple approach to track two-dimensional cell colonies composed of chemo-sensitive and resistant cell populations via fluorescence microscopy and coupled this to computational model predictions. Specifically, we first developed multiple 4T1 breast cancer cell lines resistant to varying concentrations of doxorubicin, and demonstrated how heterogeneous populations expand in a two-dimensional colony. We subjected cell populations to varied dose and frequency of chemotherapy and measured colony growth. We then built a mathematical model to describe the dynamics of both chemosensitive and chemoresistant populations, where we determined which number of doses can produce the smallest tumor size based on parameters in the system. Finally, using an in vitro model we demonstrated multiple doses can decrease overall colony growth as compared to a single dose at the same total dose. In the future, this system can be adapted to optimize dosing strategies in the setting of heterogeneous cell types or patient derived cells with varied chemoresistance.
Collapse
Affiliation(s)
- Dhruba Deb
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Shu Zhu
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Michael J LeBlanc
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Tal Danino
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
- Data Science Institute, Columbia University, New York, NY, United States
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, United States
| |
Collapse
|
22
|
Mundo AI, Muhammad A, Balza K, Nelson CE, Muldoon TJ. Longitudinal examination of perfusion and angiogenesis markers in primary colorectal tumors shows distinct signatures for metronomic and maximum-tolerated dose strategies. Neoplasia 2022; 32:100825. [PMID: 35901621 PMCID: PMC9326335 DOI: 10.1016/j.neo.2022.100825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022]
Abstract
Metronomic chemotherapy (MET) has been developed to address the shortcomings of maximum-tolerated chemotherapy (MTD) in regard to toxicity and development of resistance mechanisms in the tumor. In colorectal cancer (CRC), MET is a promising novel strategy to treat locally advanced malignancies when used as neoadjuvant chemotherapy (NAC). However, so far there are no preclinical studies to assess the impact of MET NAC in CRC to assess the benefits and challenges of this approach. Here, we used a primary model of CRC (via azoxymethane) to analyze longitudinal changes in angiogenesis in primary tumors under MET and MTD NAC using a combination of diffuse reflectance spectroscopy and mRNA expression (via qPCR). Our results show that MET and MTD NAC lead to increased mean tissue oxygen saturation (8% and 5%, respectively) and oxyhemoglobin (15% and 10%) between weeks 2 and 5 of NAC, and that such increases are caused by distinct molecular signatures in the angiogenic program. Specifically, we find that in the MET group there is a sustained increase in Hif-1a, Aldoa, and Pgk1 expression, suggesting upregulated glycolysis, whereas MTD NAC causes a significant reduction in the expression of the aforementioned genes and of Vegf, leading to vascular remodeling in MTD-treated tumors. Taken together, this study demonstrates the ability of combined optical and molecular methodologies to provide a holistic picture of tumor response to therapy in CRC in a minimally invasive manner.
Collapse
Affiliation(s)
- Ariel I Mundo
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Abdussaboor Muhammad
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Kerlin Balza
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Christopher E Nelson
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Timothy J Muldoon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA.
| |
Collapse
|
23
|
Fante MA, Felsenstein M, Mayer S, Gerken M, Klinkhammer-Schalke M, Herr W, Vogelhuber M, Reichle A, Heudobler D. All-Oral Low-Dose Chemotherapy TEPIP is Effective and Well-Tolerated in Relapsed/Refractory Patients With Aggressive B-Cell Lymphoma. Front Oncol 2022; 12:852987. [PMID: 35619924 PMCID: PMC9127443 DOI: 10.3389/fonc.2022.852987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/07/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose Treatment options in patients (pts.) with advanced relapsed and refractory aggressive B-cell lymphoma are limited. Palliative all-oral chemotherapy regimens reduce in-patient visits and contribute to quality of life. The all-oral low-dose chemotherapy regimen TEPIP comprises the conventional chemotherapy agents trofosfamide, etoposide, procarbazine, idarubicin and prednisolone. Methods Safety and efficacy of TEPIP was evaluated in an observational retrospective, single-center study at the University Medical Center Regensburg between 2010 and 2020. Treatment with TEPIP was applied for 7 or 10 days during a 28-days period. In a subgroup of fit and therapy-motivated pts. rituximab was added. End points were overall survival (OS) and progression free survival (PFS). Adverse events ≥ CTCAE grade III were reported. Results 35 highly pre-treated pts. with aggressive B-cell lymphoma were enrolled. Median age at TEPIP start was 67 years and 85% of pts. received TEPIP as ≥ third treatment line. Overall response rate (ORR) was 23% (CR 17%). Pts. benefited from additional rituximab administration (ORR 67%) and a lower number of pre-treatments (ORR 41%). The OS was 3.3 months (m) with a 1y-OS of 25.7% and the PFS amounted to 1.3 m with a 1y-PFS of 8.8%. OS and PFS were significantly prolonged in pts. that responded to treatment or additionally received rituximab. Adverse events were mainly hematological and occurred in 49% of pts. Conclusion TEPIP was well-tolerated and induced respectable response in a difficult-to-treat patient cohort. In particular, the all-oral administration enables out-patient use with palliative intent.
Collapse
Affiliation(s)
- Matthias A Fante
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Mona Felsenstein
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Stephanie Mayer
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Michael Gerken
- Bavarian Cancer Registry, Regional Centre Regensburg, Bavarian Health and Food Safety Authority, Regensburg, Germany.,Tumor Center - Institute for Quality Management and Health Services Research, University of Regensburg, Regensburg, Germany
| | - Monika Klinkhammer-Schalke
- Bavarian Cancer Registry, Regional Centre Regensburg, Bavarian Health and Food Safety Authority, Regensburg, Germany.,Tumor Center - Institute for Quality Management and Health Services Research, University of Regensburg, Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Martin Vogelhuber
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Albrecht Reichle
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Daniel Heudobler
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany.,Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
24
|
Ma J, Wang B, Shao H, Zhang S, Chen X, Li F, Liang W. Hydrogels for localized chemotherapy of liver cancer: a possible strategy for improved and safe liver cancer treatment. Drug Deliv 2022; 29:1457-1476. [PMID: 35532174 PMCID: PMC9090357 DOI: 10.1080/10717544.2022.2070299] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The systemic drug has historically been preferred for the treatment of the majority of pathological conditions, particularly liver cancer. Indeed, this mode of treatment is associated with adverse reactions, toxicity, off-target accumulation, and rapid hepatic and renal clearance. Numerous efforts have been made to design systemic therapeutic carriers to improve retention while decreasing side effects and clearance. Following systemic medication, local administration of therapeutic agents allows for higher 'effective' doses with fewer side effects, kidney accumulation, and clearance. Hydrogels are highly biocompatible and can be used for both imaging and therapy. Hydrogel-based drug delivery approach has fewer side effects than traditional chemotherapy and can deliver drugs to tumors for a longer time. The chemical and physical flexibility of hydrogels can be used to achieve disease-induced in situ accumulation as well as subsequent drug release and hydrogel-programmed degradation. Moreover, they can act as a biocompatible depot for localized chemotherapy when stimuli-responsive carriers are administrated. Herein, we summarize the design strategies of various hydrogels used for localized chemotherapy of liver cancer and their delivery routes, as well as recent research on smart hydrogels.
Collapse
Affiliation(s)
- Jianyong Ma
- Department of General Practice, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, China
| | - Bingzhu Wang
- Internal Medicine of Integrated Traditional Chinese and Western Medicine, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Haibin Shao
- Internal Medicine of Integrated Traditional Chinese and Western Medicine, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Songou Zhang
- College of Medicine, Shaoxing University, Shaoxing, China
| | - Xiaozhen Chen
- College of Medicine, Shaoxing University, Shaoxing, China
| | - Feize Li
- Internal Medicine of Integrated Traditional Chinese and Western Medicine, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenqing Liang
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| |
Collapse
|
25
|
Krishnan V, Dharamdasani V, Bakre S, Dhole V, Wu D, Budnik B, Mitragotri S. Hyaluronic Acid Nanoparticles for Immunogenic Chemotherapy of Leukemia and T-Cell Lymphoma. Pharmaceutics 2022; 14:466. [PMID: 35214193 PMCID: PMC8874923 DOI: 10.3390/pharmaceutics14020466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 02/04/2023] Open
Abstract
Ratiometric delivery of combination chemotherapy can achieve therapeutic efficacy based on synergistic interactions between drugs. It is critical to design such combinations with drugs that complement each other and reduce cancer growth through multiple mechanisms. Using hyaluronic acid (HA) as a carrier, two chemotherapeutic agents-doxorubicin (DOX) and camptothecin (CPT)-were incorporated and tested for their synergistic potency against a broad panel of blood-cancer cell lines. The pair also demonstrated the ability to achieve immunogenic cell death by increasing the surface exposure levels of Calreticulin, thereby highlighting its ability to induce apoptosis via an alternate pathway. Global proteomic profiling of cancer cells treated with HA-DOX-CPT identified pathways that could potentially predict patient sensitivity to HA-DOX-CPT. This lays the foundation for further exploration of integrating drug delivery and proteomics in personalized immunogenic chemotherapy.
Collapse
Affiliation(s)
- Vinu Krishnan
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Vimisha Dharamdasani
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Shirin Bakre
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
| | - Ved Dhole
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
| | - Debra Wu
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Bogdan Budnik
- Mass Spectrometry Proteomics and Research Laboratory, FAS Division of Science, Harvard University, Cambridge, MA 02138, USA;
| | - Samir Mitragotri
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
26
|
Wu CF, Su PH. Hepatic arterial infusion chemotherapy for liver tumor control in a patient with liver metastasis from lung small cell carcinoma, a case report and discussion. JOURNAL OF CANCER RESEARCH AND PRACTICE 2022. [DOI: 10.4103/jcrp.jcrp_29_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
27
|
Missing data handling technique in joint modeling context. BIOMEDICAL ENGINEERING ADVANCES 2021. [DOI: 10.1016/j.bea.2021.100012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
28
|
Angiogenesis and chemotherapy resistance: optimizing chemotherapy scheduling using mathematical modeling. J Cancer Res Clin Oncol 2021; 147:2281-2299. [PMID: 34050795 PMCID: PMC8236485 DOI: 10.1007/s00432-021-03657-9] [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] [Accepted: 05/03/2021] [Indexed: 12/22/2022]
Abstract
Chemotherapy remains a widely used cancer treatment. Acquired drug resistance may greatly reduce the efficacy of treatment and means to overcome it are a topic of active discussion among researchers. One of the proposed solutions is to shift the therapeutic paradigm from complete eradication of cancer to maintenance, i.e., to treat it as a chronic disease. A concept of metronomic therapy (low chemotherapy doses applied continuously) emerged in early 2000s and was henceforth shown to offer a number of benefits, including targeting endothelial cells and reducing acquired drug resistance. Using mathematical modeling and optimal control techniques, we investigate the hypothesis that lower doses of chemotherapy are beneficial for patients. Our analysis of a mathematical model of tumor growth under angiogenic signaling proposed by Hahnfeldt et al. adapted to heterogeneous tumors treated by combined anti-angiogenic agent and chemotherapy offers insights into the effects of metronomic therapy. Firstly, assuming constant long-term drug delivery, the model suggests that the longest survival time is achieved for intermediate drug doses. Secondly, by formalizing the notion of the therapeutic target being maintenance rather than eradication, we show that in the short term, optimal chemotherapy scheduling consists mainly of a drug applied at a low dose. In conclusion, we suggest that metronomic therapy is an attractive alternative to maximum tolerated dose therapies to be investigated in experimental settings and clinical trials.
Collapse
|
29
|
Sultania M, Deo SVS, Shukla NK, Sharma A, Sahoo R, Bhasker S. Low Cost, Low Dose, Oral, Neoadjuvant Chemotherapy Protocol in Locally Advanced Borderline Oral Cancers-Feasibility Study. Indian J Surg Oncol 2021; 12:67-72. [PMID: 33814834 DOI: 10.1007/s13193-020-01247-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 10/01/2020] [Indexed: 11/25/2022] Open
Abstract
A vast majority of oral cancer patients in developing countries present in an advanced stage with borderline resectable/inoperable stage to busy resource-constrained tertiary cancer centers. Conventional chemotherapy protocols are associated with issues like toxicity, tolerance, cost, and compliance. The present study was conducted to assess the feasibility of low-cost home-based chemotherapy options. Single-arm feasibility study was done in borderline resectable/inoperable oral cancer patients. Home-based oral neoadjuvant chemotherapy consisting of oral methotrexate 15 mg/m2 once a week and oral celecoxib 200 mg twice daily for 8 weeks was used. RECIST Criteria 1.1 was used to assess response to therapy. The study included 60 patients. The mean age was 51.98 years with male predominance (80%). Fifty-five patients adhered to the treatment; the compliance rate is 91.60%. Affordability (Rs 700 per month) and tolerance to therapy was 100%, and no grade III or IV toxicity was seen. Overall, 18 patients had stable disease (32.73%), partial response was seen in 15 patients (27.27%), and the disease progressed in 22 patients (40%). At the end of 8 weeks, 26 (43.3%) patients were deemed resectable. Neoadjuvant low cost, home-based metronomic chemotherapy using oral methotrexate and celecoxib seems to be a viable option in managing advanced oral cancer in resource-constrained setups.
Collapse
Affiliation(s)
- Mahesh Sultania
- Department of Surgical Oncology, All India Institute of Medical Sciences, Bhubaneswar, Odisha 751003 India
| | - S V S Deo
- Department of Surgical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - N K Shukla
- Department of Surgical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Atul Sharma
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - R Sahoo
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - S Bhasker
- Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
30
|
Sharma A, Kataria B, Biswas B, Bakhshi S, Pushpam D. Oral metronomic chemotherapy is a cost effective alternative to pazopanib in advanced soft tissue sarcoma. J Oncol Pharm Pract 2021; 28:560-568. [PMID: 33719725 DOI: 10.1177/10781552211000113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Soft tissue sarcoma(STS) is a rare and heterogeneous group of disorders with dismal outcomes in metastatic setting. Pazopanib and oral metronomic chemotherapy (OMT) have been evaluated as therapeutic options in this cohort. MATERIALS AND METHODS We conducted a retrospective, single center study evaluating 45 patients with unresectable and/or metastatic STS, who received pazopanib or oral metronomic regimen as per instituitonal protocol between January 2013 and December 2019. An informal cost minimisation analysis was conducted for both OMT and pazopanib arms, considering equivalent outcomes for both (PFS and OS). RESULTS Median PFS in OMT and Pazopanib groups was 4.13 months and 3.53 months,respectively (HR1.31, 95% CI:0.68-2.51, p = 0.41) Only one patient in the OMT group achieved an objective response (partial response) and no objective response was noted in the pazopanib group. The incidence of grade III/IVtoxicities was higher with pazopanib than with OMT (p = 0.08). There were no toxicity related deaths noted in either arm. CONCLUSIONS Our study demonstrates that OMT have a similar progression free survival (PFS) and overall survival (OS) in metastatic STS. This study raises the possibility that OMT might be an equally efficacious and less toxic alternative to pazopanib, without compromising survival outcome especially in LMIC.
Collapse
Affiliation(s)
- Aparna Sharma
- Department of Medical Oncology, All India Medical Sciences, New Delhi, India
| | - Babita Kataria
- Department of Medical Oncology, All India Medical Sciences, New Delhi, India
| | - Bivas Biswas
- Department of Medical Oncology, Tata Memorial Centre, Kolkata, India
| | - Sameer Bakhshi
- Department of Medical Oncology, All India Medical Sciences, New Delhi, India
| | - Deepam Pushpam
- Department of Medical Oncology, All India Medical Sciences, New Delhi, India
| |
Collapse
|
31
|
Brems-Eskildsen AS, Linnet S, Danø H, Luczak A, Vestlev PM, Jakobsen EH, Neimann J, Jensen CB, Dongsgaard T, Langkjer ST. Metronomic treatment of vinorelbine with oral capecitabine is tolerable in the randomized Phase 2 study XeNa including patients with HER2 non-amplified metastatic breast cancer. Acta Oncol 2021; 60:157-164. [PMID: 33259244 DOI: 10.1080/0284186x.2020.1851045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Metronomic treatment is hypothesized to be less toxic and more effective as compared to standard maximal tolerable dosing treatment in metastatic cancer disease. MATERIAL AND METHODS We tested the metronomic treatment principle with vinorelbine in a randomized phase 2 setting combined with standard capecitabine treatment in the XeNa trial with Clinical Trials.gov identifier number: NCT0141771. 120 patients with disseminated HER2 non-amplified breast cancer were included. Randomization was between Arm A: vinorelbine 60 mg/m2 day 1 + day 8 in the first cycle followed by 80 mg/m2 day 1 + day 8 in the following cycles or Arm B: vinorelbine 50 mg three times a week. Capecitabine 1000 mg/m2 twice a day for days 1-14 was administered in both arms. RESULTS The treatment was generally well-tolerated. The response rate (RR) was 24% (arm A) versus 29% (arm B) (p = .67). The clinical benefit rate (CBR) 46.8% (arm A) versus 51.7% (arm B) (p = .72). We found a median progression-free survival (PFS) of 7.1 months (95% confidence interval [CI] 3.9-10.3) in arm A and 6.3 months (95% CI 4.1-8.5) in arm B (p = .25) whereas median overall survival (OS) was 23.3 months (95% CI 20.2-26.4) in arm A and 22.3 months (95% CI 14.3-30.3) in arm B (p = .76). CONCLUSIONS We confirmed that the combination of vinorelbine and capecitabine was well tolerated. Metronomic treatment can be used with acceptable adverse events (AEs), but we did not find significant difference in the effect compared to the standard treatment.
Collapse
Affiliation(s)
| | - Søren Linnet
- Department of Oncology, Region Hospital of West Jutland, Herning, Denmark
| | - Hella Danø
- Department of Oncology, Region Hospital in Hilleroed, Hillerod, Denmark
| | - Adam Luczak
- Department of Oncology, University Hospital of Aalborg, Aalborg, Denmark
| | | | | | - Jeppe Neimann
- Department of Oncology, University Hospital of Aarhus, Aarhus, Denmark
| | | | - Trine Dongsgaard
- Department of Oncology, Region Hospital of West Jutland, Herning, Denmark
| | | |
Collapse
|
32
|
Bhat AA, Yousuf P, Wani NA, Rizwan A, Chauhan SS, Siddiqi MA, Bedognetti D, El-Rifai W, Frenneaux MP, Batra SK, Haris M, Macha MA. Tumor microenvironment: an evil nexus promoting aggressive head and neck squamous cell carcinoma and avenue for targeted therapy. Signal Transduct Target Ther 2021; 6:12. [PMID: 33436555 PMCID: PMC7804459 DOI: 10.1038/s41392-020-00419-w] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/02/2020] [Accepted: 10/15/2020] [Indexed: 12/17/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a very aggressive disease with a poor prognosis for advanced-stage tumors. Recent clinical, genomic, and cellular studies have revealed the highly heterogeneous and immunosuppressive nature of HNSCC. Despite significant advances in multimodal therapeutic interventions, failure to cure and recurrence are common and account for most deaths. It is becoming increasingly apparent that tumor microenvironment (TME) plays a critical role in HNSCC tumorigenesis, promotes the evolution of aggressive tumors and resistance to therapy, and thereby adversely affects the prognosis. A complete understanding of the TME factors, together with the highly complex tumor-stromal interactions, can lead to new therapeutic interventions in HNSCC. Interestingly, different molecular and immune landscapes between HPV+ve and HPV-ve (human papillomavirus) HNSCC tumors offer new opportunities for developing individualized, targeted chemoimmunotherapy (CIT) regimen. This review highlights the current understanding of the complexity between HPV+ve and HPV-ve HNSCC TME and various tumor-stromal cross-talk modulating processes, including epithelial-mesenchymal transition (EMT), anoikis resistance, angiogenesis, immune surveillance, metastatic niche, therapeutic resistance, and development of an aggressive tumor phenotype. Furthermore, we summarize the recent developments and the rationale behind CIT strategies and their clinical applications in HPV+ve and HPV-ve HNSCC.
Collapse
Affiliation(s)
- Ajaz A Bhat
- Functional and Molecular Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha, Qatar
| | - Parvaiz Yousuf
- Department of Zoology, School of Life Sciences, Central University of Kashmir, Ganderbal, Jammu & Kashmir, India
| | - Nissar A Wani
- Functional and Molecular Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha, Qatar
| | - Arshi Rizwan
- Department of Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | - Shyam S Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Mushtaq A Siddiqi
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Jammu & Kashmir, India
| | - Davide Bedognetti
- Laboratory of Cancer Immunogenomics, Cancer Research Department, Sidra Medicine, Doha, Qatar
| | - Wael El-Rifai
- Department of Surgery, University of Miami, Miami, FL, USA
| | | | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.,Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mohammad Haris
- Functional and Molecular Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha, Qatar. .,Laboratory Animal Research Center, Qatar University, Doha, Qatar.
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Jammu & Kashmir, India.
| |
Collapse
|
33
|
Enhanced antitumor efficacy of bile acid-lipid complex-anchored docetaxel nanoemulsion via oral metronomic scheduling. J Control Release 2020; 328:368-394. [DOI: 10.1016/j.jconrel.2020.08.067] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 01/12/2023]
|
34
|
Improving Convergence in Therapy Scheduling Optimization: A Simulation Study. MATHEMATICS 2020. [DOI: 10.3390/math8122114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The infusion times and drug quantities are two primary variables to optimize when designing a therapeutic schedule. In this work, we test and analyze several extensions to the gradient descent equations in an optimal control algorithm conceived for therapy scheduling optimization. The goal is to provide insights into the best strategies to follow in terms of convergence speed when implementing our method in models for dendritic cell immunotherapy. The method gives a pulsed-like control that models a series of bolus injections and aims to minimize a cost a function, which minimizes tumor size and to keep the tumor under a threshold. Additionally, we introduce a stochastic iteration step in the algorithm, which serves to reduce the number of gradient computations, similar to a stochastic gradient descent scheme in machine learning. Finally, we employ the algorithm to two therapy schedule optimization problems in dendritic cell immunotherapy and contrast our method’s stochastic and non-stochastic optimizations.
Collapse
|
35
|
Pangeni R, Subedi L, Jha SK, Kweon S, Kang SH, Chang KY, Choi JU, Byun Y, Park JW. Improvements in the Oral Absorption and Anticancer Efficacy of an Oxaliplatin-Loaded Solid Formulation: Pharmacokinetic Properties in Rats and Nonhuman Primates and the Effects of Oral Metronomic Dosing on Colorectal Cancer. Int J Nanomedicine 2020; 15:7719-7743. [PMID: 33116497 PMCID: PMC7555381 DOI: 10.2147/ijn.s267424] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Objective The anticancer efficacy of orally administered chemotherapeutics is often constrained by low intestinal membrane permeability and oral bioavailability. In this context, we designed a solid oral formulation of oxaliplatin (OP), a third-generation cisplatin analog, to improve oral bioavailability and investigate its application in metronomic chemotherapy. Methods An ion-pairing complex of OP with a permeation enhancer, Nα-deoxycholyl-l-lysyl-methylester (DLM), was successfully prepared and then mixed with dispersing agents (including poloxamer 188 and Labrasol) to form the solid, amorphous oral formulation OP/DLM (OP/DLM-SF; hereafter, ODSF). Results The optimized powder formulation was sized in the nanoscale range (133±1.47 nm). The effective permeability of OP increased by 12.4-fold after ionic complex formation with DLM and was further increased by 24.0-fold after incorporation into ODSF. ODSF exhibited respective increases of 128% and 1010% in apparent permeability across a Caco-2 monolayer, compared to OP/DLM and OP. Furthermore, inhibition of bile acid transporters by actinomycin D and caveola-mediated uptake by brefeldin in Caco-2 cell monolayers reduced the apparent permeability values of ODSF by 58.4% and 51.1%, respectively, suggesting predominant roles for bile acid transporters and caveola-mediated transport in intestinal absorption of ODSF. In addition, macropinocytosis and paracellular and transcellular passive transport significantly influenced the intestinal permeation of ODSF. The oral bioavailabilities of ODSF in rats and monkeys were 68.2% and 277% higher, respectively, than the oral bioavailability of free OP. In vivo analyses of anticancer efficacy in CT26 and HCT116 cell-bearing mice treated with ODSF demonstrated significant suppression of tumor growth, with respective maximal tumor volume reductions of 7.77-fold and 4.07-fold, compared to controls. Conclusion ODSF exhibits therapeutic potential, constituting an effective delivery system that increases oral bioavailability, with applications to metronomic chemotherapy.
Collapse
Affiliation(s)
- Rudra Pangeni
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan-gun, Jeonnam, 58554, Republic of Korea
| | - Laxman Subedi
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Mokpo National University, Muan-gun, Jeonnam, 58554, Republic of Korea
| | - Saurav Kumar Jha
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Mokpo National University, Muan-gun, Jeonnam, 58554, Republic of Korea
| | - Seho Kweon
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seo-Hee Kang
- Global R&D Center, IcureBNP, Seoul 08511, Republic of Korea
| | | | - Jeong Uk Choi
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Youngro Byun
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin Woo Park
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan-gun, Jeonnam, 58554, Republic of Korea.,Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Mokpo National University, Muan-gun, Jeonnam, 58554, Republic of Korea
| |
Collapse
|
36
|
Sarangi SC, Sopory P, Pattnaik SS, Reeta KH. Antibody-drug conjugates, cancer immunotherapy, and metronomic chemotherapy as novel approaches in cancer management. Indian J Pharmacol 2020; 52:402-413. [PMID: 33283772 PMCID: PMC8025760 DOI: 10.4103/ijp.ijp_475_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/19/2019] [Accepted: 10/15/2020] [Indexed: 01/04/2023] Open
Abstract
Treatment of cancer is a major challenge even though the pathophysiology is becoming clearer with time. A number of new chemical entities are developed to target cancer growth inhibition, but the targeted delivery of these products still needs novel research. This is of utmost importance not only for higher efficacy but also for a reduction in systemic toxicity and cost of treatment. Although multiple novel targets and molecules are being researched, most of them could not pass the regulatory approval process, due to low benefit-risk ratio and lack of target specificity. Failure of a majority of these drugs was in part due to their superiority claimed via surrogate markers. Despite these, currently, more than 100 chemotherapeutic agents are in practice. This review paper discusses in detail the molecular basis, drug discovery, and pros and cons over conventional treatment approaches of three novel approaches in cancer therapy, i.e., (i) antibody-drug conjugates, (ii) cancer immunotherapy, and (iii) metronomic chemotherapy. All the drugs developed using these three novel approaches were compared against the established treatment regimens in clinical trials with clinical end points, such as overall survival, progression-free survival, and quality of life.
Collapse
Affiliation(s)
| | - Pranav Sopory
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | | | - KH Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
37
|
Reynolds BA, Oli MW, Oli MK. Eco-oncology: Applying ecological principles to understand and manage cancer. Ecol Evol 2020; 10:8538-8553. [PMID: 32884638 PMCID: PMC7452771 DOI: 10.1002/ece3.6590] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/25/2022] Open
Abstract
Cancer is a disease of single cells that expresses itself at the population level. The striking similarities between initiation and growth of tumors and dynamics of biological populations, and between metastasis and ecological invasion and community dynamics suggest that oncology can benefit from an ecological perspective to improve our understanding of cancer biology. Tumors can be viewed as complex, adaptive, and evolving systems as they are spatially and temporally heterogeneous, continually interacting with each other and with the microenvironment and evolving to increase the fitness of the cancer cells. We argue that an eco-evolutionary perspective is essential to understand cancer biology better. Furthermore, we suggest that ecologically informed therapeutic approaches that combine standard of care treatments with strategies aimed at decreasing the evolutionary potential and fitness of neoplastic cells, such as disrupting cell-to-cell communication and cooperation, and preventing successful colonization of distant organs by migrating cancer cells, may be effective in managing cancer as a chronic condition.
Collapse
Affiliation(s)
- Brent A. Reynolds
- Department of NeurosurgeryCollege of MedicineUniversity of FloridaGainesvilleFLUSA
| | - Monika W. Oli
- Department of Microbiology and Cell ScienceInstitute of Food and Agricultural SciencesUniversity of FloridaGainesvilleFLUSA
| | - Madan K. Oli
- Department of Wildlife Ecology and ConservationInstitute of Food and Agricultural SciencesUniversity of FloridaGainesvilleFLUSA
| |
Collapse
|
38
|
Lee JJ, Liao AT, Wang SL. Outcome of Canine Multicentric Lymphoma after Single or Divided Treatment with Cyclophosphamide in Multidrug Chemotherapy. Top Companion Anim Med 2020; 41:100461. [PMID: 32823160 DOI: 10.1016/j.tcam.2020.100461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 10/24/2022]
Abstract
Cyclophosphamide is commonly used in combination chemotherapy to treat dogs with lymphoma. The metabolite of cyclophosphamide, acrolein, can irritate urinary bladder and cause sterile hemorrhagic cystitis. Dividing the administration of cyclophosphamide across multiple days may reduce the concentration of this metabolite in urinary bladder and reduce the possibility of cystitis. However, the impact of the therapeutic effect of this modification is not evaluated and compared to traditional single maximum-tolerated dose regimen. Seventy-two dogs with multicentric lymphoma received either bolus doses or divided doses of cyclophosphamide were included in this study. The incidence of hemorrhagic cystitis between 2 cyclophosphamide treatment groups was not significantly different (P = .357). There was no statistical difference in progression-free survival and survival time between 2 groups (P = .267 and P = .346). This modification of cyclophosphamide administration did not reduce the side effect of cystitis or affect remission and survival times in lymphoma dogs.
Collapse
Affiliation(s)
- Jih-Jong Lee
- Graduate Institute of Veterinary Clinical Science, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan; Animal Cancer Center, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan; National Taiwan University Veterinary Hospital, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Albert Taiching Liao
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan; Animal Cancer Center, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Shang-Lin Wang
- Graduate Institute of Veterinary Clinical Science, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan; Animal Cancer Center, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan; National Taiwan University Veterinary Hospital, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
39
|
Multiple low dose therapy as an effective strategy to treat EGFR inhibitor-resistant NSCLC tumours. Nat Commun 2020; 11:3157. [PMID: 32572029 PMCID: PMC7308397 DOI: 10.1038/s41467-020-16952-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 06/04/2020] [Indexed: 12/31/2022] Open
Abstract
Resistance to targeted cancer drugs is thought to result from selective pressure exerted by a high drug dose. Partial inhibition of multiple components in the same oncogenic signalling pathway may add up to complete pathway inhibition, while decreasing the selective pressure on each component to acquire a resistance mutation. We report here testing of this Multiple Low Dose (MLD) therapy model in EGFR mutant NSCLC. We show that as little as 20% of the individual effective drug doses is sufficient to completely block MAPK signalling and proliferation when used in 3D (RAF + MEK + ERK) or 4D (EGFR + RAF + MEK + ERK) inhibitor combinations. Importantly, EGFR mutant NSCLC cells treated with MLD therapy do not develop resistance. Using several animal models, we find durable responses to MLD therapy without associated toxicity. Our data support the notion that MLD therapy could deliver clinical benefit, even for those having acquired resistance to third generation EGFR inhibitor therapy. A drug used at the maximum tolerated dose can exert a strong selective pressure on cancer cells leading to resistance. In this study, the authors demonstrate the efficacy of using low dose of multiple drugs for preventing and treating resistance to EGFR tyrosine kinase inhibitors in NSCLC cells.
Collapse
|
40
|
Optimal Control Theory for Personalized Therapeutic Regimens in Oncology: Background, History, Challenges, and Opportunities. J Clin Med 2020; 9:jcm9051314. [PMID: 32370195 PMCID: PMC7290915 DOI: 10.3390/jcm9051314] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
Optimal control theory is branch of mathematics that aims to optimize a solution to a dynamical system. While the concept of using optimal control theory to improve treatment regimens in oncology is not novel, many of the early applications of this mathematical technique were not designed to work with routinely available data or produce results that can eventually be translated to the clinical setting. The purpose of this review is to discuss clinically relevant considerations for formulating and solving optimal control problems for treating cancer patients. Our review focuses on two of the most widely used cancer treatments, radiation therapy and systemic therapy, as they naturally lend themselves to optimal control theory as a means to personalize therapeutic plans in a rigorous fashion. To provide context for optimal control theory to address either of these two modalities, we first discuss the major limitations and difficulties oncologists face when considering alternate regimens for their patients. We then provide a brief introduction to optimal control theory before formulating the optimal control problem in the context of radiation and systemic therapy. We also summarize examples from the literature that illustrate these concepts. Finally, we present both challenges and opportunities for dramatically improving patient outcomes via the integration of clinically relevant, patient-specific, mathematical models and optimal control theory.
Collapse
|
41
|
Scharovsky OG, Rico MJ, Mainetti LE, Perroud HA, Rozados VR. Achievements and challenges in the use of metronomics for the treatment of breast cancer. Biochem Pharmacol 2020; 175:113909. [DOI: 10.1016/j.bcp.2020.113909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022]
|
42
|
Al‐Zeheimi N, Adham SA. Modeling Neoadjuvant chemotherapy resistance in vitro increased NRP-1 and HER2 expression and converted MCF7 breast cancer subtype. Br J Pharmacol 2020; 177:2024-2041. [PMID: 31883395 PMCID: PMC7161552 DOI: 10.1111/bph.14966] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 11/05/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Patients with locally advanced breast cancer usually receive third-generation neoadjuvant chemotherapy (NAC). Although NAC treatment improved the overall survival, patients' response varies, some acquire resistance and others exhibit a conversion in their breast cancer molecular subtype. We aimed to identify the molecular changes involved in NAC resistance attempting to find new therapeutic targets in different breast cancer subtypes. EXPERIMENTAL APPROACH We modelled NAC treatments used in clinical practice and generated resistant cell lines in vitro. The resistant cells were generated by consecutive treatment with four cycles of doxorubicin (adriamycin)/cyclophosphamide (4xAC) followed by an additional four cycles of paclitaxel (4xAC + 4xPAC). KEY RESULTS Our data revealed distinct mechanisms of resistance depending on breast cancer subtype and drugs used. MDA-MB-231 cells resistant to 4xAC + 4xPAC activated neuropilin-1/TNC/integrin β3/FAK/NF-κBp65 axis and displayed a decrease in breast cancer resistance protein (BCRP/ABCB2). However, MCF7 cells resistant to 4xAC treatments induced HER2 expression, which converted MCF7 subtype from luminal A to luminal B HER2 type, up-regulated neuropilin-1, oestrogen receptor-α, and EGFR, and activated PI3K/Akt/NF-κBp65 axis. However, MCF7 cells resistant to 4xAC + 4xPAC exhibited down-regulation of the survival axis and up-regulated BCRP/ABCG2. Co-immunoprecipitation demonstrated a novel interaction between HER2 and neuropilin-1 driving the resistance features. CONCLUSIONS AND IMPLICATIONS The concurrent increase in neuropilin-1 and HER2 upon resistance and the inverse relationship between neuropilin-1 and BCRP/ABCG2 suggest that, in addition to HER2, neuropilin-1 status should be assessed in patients undergoing NAC, and as a potential drug target for refractory breast cancer.
Collapse
Affiliation(s)
- Noura Al‐Zeheimi
- Department of Biology, College of ScienceSultan Qaboos UniversityMuscatOman
| | - Sirin A. Adham
- Department of Biology, College of ScienceSultan Qaboos UniversityMuscatOman
| |
Collapse
|
43
|
Zhong H, Lai Y, Zhang R, Daoud A, Feng Q, Zhou J, Shang J. Low Dose Cyclophosphamide Modulates Tumor Microenvironment by TGF-β Signaling Pathway. Int J Mol Sci 2020; 21:ijms21030957. [PMID: 32023984 PMCID: PMC7038197 DOI: 10.3390/ijms21030957] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 12/24/2022] Open
Abstract
The tumor microenvironment has been recently recognized as a critical contributor to cancer progression and anticancer therapy-resistance. Cyclophosphamide (CTX) is a cytotoxic agent commonly used in clinics for the treatment of cancer. Previous reports demonstrated that CTX given at low continuous doses, known as metronomic schedule, mainly targets endothelial cells and circulating Tregs with unknown mechanisms. Here, we investigated the antitumor activity of two different metronomic schedules of CTX along with their corresponding MTD regimen and further explored their effect on immune function and tumor microenvironment. Toxicity evaluation was monitored by overall survival rate, weight loss, and histopathological analysis. A nude mouse model of Lewis lung cancer was established to assess the anti-metastatic effects of CTX in vivo. CD4+, CD8+, and CD4+CD25+FoxP3 T cells were selected by flow cytometry analysis. Low and continuous administration of CTX was able to restore immune function via increase of CD4+/CD8+ T cells and depletion of T regulatory cells, not only in circulatory and splenic compartments, but also at the tumor site. Low-dose CTX also reduced myofibroblasts, accompanied with an increased level of E-cadherin and low N-cadherin, both in the primary tumor and lung through the TGF-β pathway by the downregulated expression of TGF-β receptor 2. Our data may indicate that several other molecular mechanisms of CTX for tumor may be involved in metronomic chemotherapy, besides targeting angiogenesis and regulatory T cells.
Collapse
Affiliation(s)
- Hui Zhong
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (Y.L.); (R.Z.); (Q.F.)
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China;
| | - Yifan Lai
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (Y.L.); (R.Z.); (Q.F.)
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China;
| | - Rui Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (Y.L.); (R.Z.); (Q.F.)
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China;
| | - Abdelkader Daoud
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China;
| | - Qingyuan Feng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (Y.L.); (R.Z.); (Q.F.)
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China;
| | - Jia Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (Y.L.); (R.Z.); (Q.F.)
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China;
- Correspondence: (J.Z.); (J.S.); Tel./Fax: +86-25-83271142 (J.S.)
| | - Jing Shang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (Y.L.); (R.Z.); (Q.F.)
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China;
- Correspondence: (J.Z.); (J.S.); Tel./Fax: +86-25-83271142 (J.S.)
| |
Collapse
|
44
|
Arthur C, Jeffrey A, Yip E, Katsioulas V, Nalpantidis A, Kerridge I, Greenwood M, Coyle L, Mackinlay N, Fay K, Enjeti A, Shortt J, Stevenson W. Prolonged administration of low-dose cytarabine and thioguanine in elderly patients with acute myeloid leukaemia (AML) achieves high complete remission rates and prolonged survival. Leuk Lymphoma 2019; 61:831-839. [PMID: 31809629 DOI: 10.1080/10428194.2019.1697876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The prognosis of AML in elderly patients is poor and research into novel therapeutic approaches is urgently needed. This study examined the use of low-dose chemotherapy with cytarabine and thioguanine administered in repetitive cycles in 62 elderly patients with newly diagnosed or relapsed/refractory AML. The overall response rate was 58% in the total cohort. Response rates (CR/CRi) were significantly higher in patients with newly diagnosed AML (74%) compared to patients with relapsed/refractory disease (25%, p = .0004). Kaplan-Meier estimate of overall survival was 289 days (95% CI; 183-395 days) with a relapse rate of 65.7%. The induction mortality rate was 16.1% with treatment successfully undertaken in the outpatient setting. Similar clinical outcomes were observed in a retrospective analysis of a second cohort of 25 AML patients treated at a different site. These results support the use of a sustained low intensity chemotherapy approach as a therapeutic option for elderly patients with AML.
Collapse
Affiliation(s)
- Christopher Arthur
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia
| | - Anthony Jeffrey
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia
| | - Eva Yip
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia
| | - Vicki Katsioulas
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia
| | | | - Ian Kerridge
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia.,Department of Medicine, University of Sydney, Sydney, Australia
| | - Matthew Greenwood
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia.,Department of Medicine, University of Sydney, Sydney, Australia
| | - Luke Coyle
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia
| | - Naomi Mackinlay
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia
| | - Keith Fay
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia
| | - Anoop Enjeti
- Department of Haematology, Pathology North, John Hunter Hospital, Newcastle, Australia
| | - Jake Shortt
- Department of Clinical Haematology, Monash Health, Clayton, Australia.,School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
| | - William Stevenson
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia.,Department of Medicine, University of Sydney, Sydney, Australia
| |
Collapse
|
45
|
Tafuto S, von Arx C, Capozzi M, Tatangelo F, Mura M, Modica R, Barretta ML, Di Sarno A, Tornesello ML, Colao A, Ottaiano A. Safety and Activity of Metronomic Temozolomide in Second-Line Treatment of Advanced Neuroendocrine Neoplasms. J Clin Med 2019; 8:1224. [PMID: 31443197 PMCID: PMC6723560 DOI: 10.3390/jcm8081224] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/07/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Platinum-based chemotherapy is the mainstay of front-line treatment of patients affected by pluri-metastatic intermediate/high grade NeuroEndocrine Neoplasms (NENs). However, there are no standard second-line treatments at disease progression. Previous clinical experiences have evidenced that temozolomide (TMZ), an oral analog of dacarbazine, is active against NENs at standard doses of 150 to 200 mg/mq per day on days 1 to 5 of a 28-day cycle, even if a significant treatment-related toxicity is reported. METHODS Metastatic NENs patients were treated at the ENETS (European NeuroEndocrine Tumor Society) center of excellence of Naples (Italy), from 2014 to 2017 with a second-line alternative metronomic schedule of TMZ, 75 mg/m2 per os "one week on/one week off". Toxicity was graded with NCI-CTC criteria v4.0; objective responses with RECIST v1.1 and performance status (PS) according to ECOG. RESULTS Twenty-six consecutive patients were treated. Median age was 65.5 years. The predominant primary organs were pancreas and lung. Grading was G2 in 11 patients, G3 in 15. More than half of patients had a PS 2 (15 vs. 11 with PS 1). The median time-on-temozolomide therapy was 12.2 months (95% CI: 11.4-19.6). No G3/G4 toxicities were registered. Complete response was obtained in 1 patient, partial response in 4, stable disease in 19 (disease control rate: 92.3%), and progressive disease in 2. The median overall survival from TMZ start was 28.3 months. PS improved in 73% of patients. CONCLUSIONS Metronomic TMZ is a suitable treatment for G2 and G3 NENs particularly in PS 2 patients. Prospective and larger trials are needed to confirm these results.
Collapse
Affiliation(s)
- Salvatore Tafuto
- Department of Abdominal Oncology, Istituto Nazionale Tumori, IRCCS-Fondazione "G. Pascale", 80131 Naples, Italy
- ENETs (European NeuroEndocrine Tumors Society), Center of Excellence of Naples, 80131 Naples, Italy
| | - Claudia von Arx
- Department of Abdominal Oncology, Istituto Nazionale Tumori, IRCCS-Fondazione "G. Pascale", 80131 Naples, Italy
- ENETs (European NeuroEndocrine Tumors Society), Center of Excellence of Naples, 80131 Naples, Italy
- Department of Surgery and Cancer, Imperial College London, London W12 0HS, UK
| | - Monica Capozzi
- Department of Abdominal Oncology, Istituto Nazionale Tumori, IRCCS-Fondazione "G. Pascale", 80131 Naples, Italy.
- ENETs (European NeuroEndocrine Tumors Society), Center of Excellence of Naples, 80131 Naples, Italy.
| | - Fabiana Tatangelo
- ENETs (European NeuroEndocrine Tumors Society), Center of Excellence of Naples, 80131 Naples, Italy
- Department of Pathology, Istituto Nazionale Tumori, IRCCS-Fondazione "G. Pascale", 80131 Naples, Italy
| | - Manuela Mura
- Department of Surgery and Cancer, Imperial College London, London W12 0HS, UK
- ENETs (European NeuroEndocrine Tumors Society), Center of Excellence of Naples, 80131 Naples, Italy
| | - Roberta Modica
- ENETs (European NeuroEndocrine Tumors Society), Center of Excellence of Naples, 80131 Naples, Italy
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Maria Luisa Barretta
- ENETs (European NeuroEndocrine Tumors Society), Center of Excellence of Naples, 80131 Naples, Italy
- UOC of Radiology, Istituto Nazionale Tumori, IRCCS-Fondazione "G. Pascale", 80131 Naples, Italy
| | - Antonella Di Sarno
- ENETs (European NeuroEndocrine Tumors Society), Center of Excellence of Naples, 80131 Naples, Italy
- UOC of Oncology, A.O. dei Colli, Monaldi Unit, 80131 Naples, Italy
| | - Maria Lina Tornesello
- ENETs (European NeuroEndocrine Tumors Society), Center of Excellence of Naples, 80131 Naples, Italy
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", 80131 Naples, Italy
| | - Annamaria Colao
- ENETs (European NeuroEndocrine Tumors Society), Center of Excellence of Naples, 80131 Naples, Italy
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Alessandro Ottaiano
- Department of Abdominal Oncology, Istituto Nazionale Tumori, IRCCS-Fondazione "G. Pascale", 80131 Naples, Italy
- ENETs (European NeuroEndocrine Tumors Society), Center of Excellence of Naples, 80131 Naples, Italy
| |
Collapse
|
46
|
Langkjer ST, Kenholm J, Jensen JD, Wedervang K, Brixen AT, Grunnet M, Stenbygaard L, Gilje B, Danø H, Glavicic V, Jacobsen EH, Brems-Eskildsen AS, Kruse HL, Dongsgaard T, Neimann J, Geisler J. The NAME trial: a direct comparison of classical oral Navelbine versus Metronomic Navelbine in metastatic breast cancer. Future Oncol 2019; 15:2561-2569. [DOI: 10.2217/fon-2019-0124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Chemotherapy for metastatic breast cancer (MBC) is in general given in cycles of maximum tolerated doses to potentially maximize the therapeutic outcome. However, when compared with targeted therapies for MBC, conventional and dose intensified chemotherapy has caused only modest survival benefits during the recent decades, often compromising the quality of life considerably. Navelbine is an antineoplastic agent that has shown efficacy in the treatment of a variety of cancer types, including breast cancer. Early clinical trials involving both breast cancer and lung cancer patients suggest that metronomic dosing of Navelbine might be at least as effective as classical administration (once weekly, etc.). The NAME trial compares these two strategies of Navelbine administration in MBC patients.
Collapse
Affiliation(s)
- Sven Tyge Langkjer
- Department of Oncology, Aarhus University Hospital, 8200 Aarhus C, Denmark
| | - Julia Kenholm
- Department of Oncology, Regionshospitalet Herning, 7400 Herning, Denmark
| | | | - Kim Wedervang
- Department of Oncology, Sønderborg Sygehus, 6400 Sønderborg, Denmark
| | | | - Mie Grunnet
- Department of Oncology, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Lars Stenbygaard
- Department of Oncology, Aalborg Sygehus Syd, 9100 Aalborg, Denmark
| | - Bjørnar Gilje
- Department of Hematology & Oncology, Stavanger University Hospital, 4011 Stavanger, Norway
| | - Hella Danø
- Department of Oncology, Hilleroed Hospital, 3400 Hilleroed, Denmark
| | - Vesna Glavicic
- Department of Oncology, Naestved, 4700 Naestved, Denmark
| | | | | | - Helle Lemvig Kruse
- Department of Oncology, Aarhus University Hospital, 8200 Aarhus C, Denmark
| | - Trine Dongsgaard
- Department of Oncology, Regionshospitalet Herning, 7400 Herning, Denmark
| | - Jeppe Neimann
- Department of Oncology, Aarhus University Hospital, 8200 Aarhus C, Denmark
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| |
Collapse
|
47
|
Alessandri G, Coccè V, Pastorino F, Paroni R, Dei Cas M, Restelli F, Pollo B, Gatti L, Tremolada C, Berenzi A, Parati E, Brini AT, Bondiolotti G, Ponzoni M, Pessina A. Microfragmented human fat tissue is a natural scaffold for drug delivery: Potential application in cancer chemotherapy. J Control Release 2019; 302:2-18. [DOI: 10.1016/j.jconrel.2019.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 03/10/2019] [Accepted: 03/15/2019] [Indexed: 12/16/2022]
|
48
|
Lajous H, Lelièvre B, Vauléon E, Lecomte P, Garcion E. Rethinking Alkylating(-Like) Agents for Solid Tumor Management. Trends Pharmacol Sci 2019; 40:342-357. [PMID: 30979523 DOI: 10.1016/j.tips.2019.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/15/2019] [Accepted: 03/06/2019] [Indexed: 12/20/2022]
Abstract
Although old molecules, alkylating agents and platinum derivatives are still widely used in the treatment of various solid tumors. However, systemic toxicity and cellular resistance mechanisms impede their efficacy. Innovative strategies, including local administration, optimization of treatment schedule/dosage, synergistic combinations, and the encapsulation of bioactive molecules in smart, multifunctional drug delivery systems, have shown promising results in potentiating anticancer activity while circumventing such hurdles. Furthermore, questioning of the old paradigm according to which nuclear DNA is the critical target of their anticancer activity has shed light on subcellular alternative and neglected targets that obviously participate in the mediation of cytotoxicity or resistance. Thus, rethinking of the use of these pivotal antineoplastic agents appears critical to improve clinical outcomes in the management of solid tumors.
Collapse
Affiliation(s)
- Hélène Lajous
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France; Center for Education and Research on Macromolecules (CERM), CESAM Research Unit, University of Liege, B6a Sart-Tilman, B-4000 Liege, Belgium
| | - Bénédicte Lelièvre
- Centre Régional de Pharmacovigilance, Laboratoire de Pharmacologie-Toxicologie, CHU Angers, 4 rue Larrey, F-49100 Angers, France
| | - Elodie Vauléon
- Centre Eugène Marquis, Rennes, France; INSERM U1242, Université de Rennes 1, Rennes, France
| | - Philippe Lecomte
- Center for Education and Research on Macromolecules (CERM), CESAM Research Unit, University of Liege, B6a Sart-Tilman, B-4000 Liege, Belgium; Equivalent contribution
| | - Emmanuel Garcion
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France; Equivalent contribution.
| |
Collapse
|
49
|
Demeure MJ. The Role of Precision Medicine in the Diagnosis and Treatment of Patients with Rare Cancers. Cancer Treat Res 2019; 178:81-108. [PMID: 31209842 DOI: 10.1007/978-3-030-16391-4_3] [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: 06/09/2023]
Abstract
Rare cancers pose unique challenges for patients and their physicians arising from a lack of information regarding the best therapeutic options. Very often, a lack of clinical trial data leads physicians to choose treatments based on small case series or case reports. Precision medicine based on genomic analysis of tumors may allow for selection of better treatments with greater efficacy and less toxicity. Physicians are increasingly using genetics to identify patients at high risk for certain cancers to allow for early detection or prophylactic interventions. Genomics can be used to inform prognosis and more accurately establish a diagnosis. Genomic analysis may also expose therapeutic targets for which drugs are currently available and approved for use in other cancers. Notable successes in the treatment of previously refractory cancers have resulted. New more advanced sequencing technologies, tools for interpretation, and an increasing array of targeted drugs offer additional hope, but challenges remain.
Collapse
Affiliation(s)
- Michael J Demeure
- Hoag Family Cancer Institute, Newport Beach, CA, USA.
- Translational Genomics Research Institute, Phoenix, AZ, USA.
| |
Collapse
|
50
|
High-resolution deconstruction of evolution induced by chemotherapy treatments in breast cancer xenografts. Sci Rep 2018; 8:17937. [PMID: 30560892 PMCID: PMC6298990 DOI: 10.1038/s41598-018-36184-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/14/2018] [Indexed: 12/30/2022] Open
Abstract
The processes by which tumors evolve are essential to the efficacy of treatment, but quantitative understanding of intratumoral dynamics has been limited. Although intratumoral heterogeneity is common, quantification of evolution is difficult from clinical samples because treatment replicates cannot be performed and because matched serial samples are infrequently available. To circumvent these problems we derived and assayed large sets of human triple-negative breast cancer xenografts and cell cultures from two patients, including 86 xenografts from cyclophosphamide, doxorubicin, cisplatin, docetaxel, or vehicle treatment cohorts as well as 45 related cell cultures. We assayed these samples via exome-seq and/or high-resolution droplet digital PCR, allowing us to distinguish complex therapy-induced selection and drift processes among endogenous cancer subclones with cellularity uncertainty <3%. For one patient, we discovered two predominant subclones that were granularly intermixed in all 48 co-derived xenograft samples. These two subclones exhibited differential chemotherapy sensitivity–when xenografts were treated with cisplatin for 3 weeks, the post-treatment volume change was proportional to the post-treatment ratio of subclones on a xenograft-to-xenograft basis. A subsequent cohort in which xenografts were treated with cisplatin, allowed a drug holiday, then treated a second time continued to exhibit this proportionality. In contrast, xenografts from other treatment cohorts, spatially dissected xenograft fragments, and cell cultures evolved in diverse ways but with substantial population bottlenecks. These results show that ecosystems susceptible to successive retreatment can arise spontaneously in breast cancer in spite of a background of irregular subclonal bottlenecks, and our work provides to our knowledge the first quantification of the population genetics of such a system. Intriguingly, in such an ecosystem the ratio of common subclones is predictive of the state of treatment susceptibility, showing how measurements of subclonal heterogeneity could guide treatment for some patients.
Collapse
|