1
|
Dommer AP, Kumarasamy V, Wang J, O’Connor TN, Roti M, Mahan S, McLean K, Knudsen ES, Witkiewicz AK. Tumor Suppressors Condition Differential Responses to the Selective CDK2 Inhibitor BLU-222. Cancer Res 2025; 85:1310-1326. [PMID: 39945638 PMCID: PMC11977231 DOI: 10.1158/0008-5472.can-24-2244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/15/2024] [Accepted: 01/22/2025] [Indexed: 03/23/2025]
Abstract
Cyclin-dependent kinase 2 (CDK2) inhibitors have recently been developed and have entered clinical trials. Combination approaches can help broaden the use of therapeutic agents and establish more effective treatments. Here, we evaluated the selective CDK2 inhibitor BLU-222 for mechanisms of response in the context of ovarian and breast cancer models. Sensors of cellular CDK activity indicated that sensitivity to either CDK4/6 or CDK2 inhibition was related to the differential dependence on a single CDK for G1-S transition. Unlike CDK4/6 inhibitors, BLU-222 was able to robustly inhibit proliferation through cell-cycle inhibition in both G1 and G2 phases. However, it remained possible for cells to reenter the cell cycle upon drug withdrawal. The antiproliferative strength and impact on G1-S versus G2-M accumulation was mediated by the RB tumor suppressor. To broaden the sensitivity to CDK2 inhibition, combinatorial drug screens were performed that identified both synergistic (e.g., CDK4/6 inhibitors) and antagonistic (e.g., WEE1 inhibitors) relationships. Models that were exceptionally sensitive to CDK2 inhibition displayed coordinate expression of cyclin E1 and P16INK4A, an endogenous CDK4/6 inhibitor. Functional studies demonstrated that P16INK4A and CDK4/6 activity were key mediators of sensitivity to BLU-222. Clinical gene and protein expression analyses revealed a positive correlation between cyclin E1 and P16INK4A and identified that ∼25% of ovarian cancers exhibited coordinate expression of cyclin E, P16INK4A, and RB, indicative of strong sensitivity to CDK2 inhibition. Together, this work advances a precision strategy for the use of CDK2 inhibitors in the context of ovarian and breast cancers. Significance: The CDK2-specific inhibitor BLU-222 shows preclinical efficacy in breast and ovarian cancer with select determinants of response and holds promise in combinatorial strategies. See related article by House and colleagues, p. 1297.
Collapse
Affiliation(s)
- Adam P. Dommer
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Vishnu Kumarasamy
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jianxin Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Thomas N. O’Connor
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Michelle Roti
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sidney Mahan
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Karen McLean
- Department of Gynecology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Erik S. Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Agnieszka K. Witkiewicz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
2
|
Giri S, Lamichhane G, Pandey J, Khadayat R, K. C. S, Devkota HP, Khadka D. Immune Modulation and Immunotherapy in Solid Tumors: Mechanisms of Resistance and Potential Therapeutic Strategies. Int J Mol Sci 2025; 26:2923. [PMID: 40243502 PMCID: PMC11989189 DOI: 10.3390/ijms26072923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Understanding the modulation of specific immune cells within the tumor microenvironment (TME) offers new hope in cancer treatments, especially in cancer immunotherapies. In recent years, immune modulation and resistance to immunotherapy have become critical challenges in cancer treatments. However, novel strategies for immune modulation have emerged as promising approaches for oncology due to the vital roles of the immunomodulators in regulating tumor progression and metastasis and modulating immunological responses to standard of care in cancer treatments. With the progress in immuno-oncology, a growing number of novel immunomodulators and mechanisms are being uncovered, offering the potential for enhanced clinical immunotherapy in the near future. Thus, gaining a comprehensive understanding of the broader context is essential. Herein, we particularly summarize the paradoxical role of tumor-related immune cells, focusing on how targeted immune cells and their actions are modulated by immunotherapies to overcome immunotherapeutic resistance in tumor cells. We also highlight the molecular mechanisms employed by tumors to evade the long-term effects of immunotherapeutic agents, rendering them ineffective.
Collapse
Affiliation(s)
- Suman Giri
- Asian College for Advance Studies, Purbanchal University, Satdobato, Lalitpur 44700, Nepal;
| | - Gopal Lamichhane
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Jitendra Pandey
- Department of Chemistry, University of Hawai’i at Manoa, 2545 McCarthy Mall, Honolulu, HI 96822, USA;
| | - Ramesh Khadayat
- Patan Hospital, Patan Academic of Health Sciences, Lagankhel, Lalitpur 44700, Nepal;
| | - Sindhu K. C.
- Department of Pharmacology, Chitwan Medical College, Tribhuwan University, Bharatpur-05, Chitwan 44200, Nepal;
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Oehonmachi 5-1, Chuo-ku, Kumamoto 862-0973, Japan;
- Headquarters for Admissions and Education, Kumamoto University, Kurokami, 2-39-1, Chuo-ku, Kumamoto 860-8555, Japan
| | - Dipendra Khadka
- NADIANBIO Co., Ltd., Wonkwang University School of Medicine, Business Incubation Center R201-1, Iksan 54538, Jeonbuk, Republic of Korea
- KHAS Health Pvt. Ltd., Dhangadhi-04, Kailali 10910, Nepal
| |
Collapse
|
3
|
Kaga E, Kaga S, Altunbas K, Okumus N. Inhalable Nano Formulation of Cabazitaxel: A Comparative Study with Intravenous Route. Macromol Biosci 2025:e2400567. [PMID: 39888152 DOI: 10.1002/mabi.202400567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/25/2024] [Indexed: 02/01/2025]
Abstract
Chemotherapy is generally given by intravenous (IV) administration which provides higher bioavailability than other systemic routes. However, in the case of lung cancer, the pulmonary (INH) route is the other choice for inhalable formulations. In the study, biochemical and histological parameters of Cabazitaxel (CBZ) free (2 mg kg-1) and nanoparticle (NP) (2 mg kg-1 CBZ equivalent) formulations are investigated after IV and INH administration in rats. The nanoformulation of CBZ is obtained using PEGylated polystyrene (PEG-PST) nanoparticles obtained by PISA. While a nose and head-only device is used for INH administration, a jugular vein is used as the IV route. Blood samples (blank, 24 h, and 48 h) are collected via carotid artery cannulas without handling in metabolism cages. According to biochemical parameters, free CBZ formulation applied via IV or INH route shows higher systemic toxicity. On the other hand, the nanoformulation of CBZ showed no signs of toxicity in both IV or INH routes. Higher and longer retention is observed in the case of inhaled nanoformulation. Histological analysis showed higher alveolar macrophage migration for inhaled nanoformulation due to enhanced retention. Results showed that nanotechnology and the lung defense system gave the advantage to end up with an inhalable nanomedicine formulation for lung cancer.
Collapse
Affiliation(s)
- Elif Kaga
- Department of Medical Services and Techniques, Afyonkarahisar Health Sciences University, Afyonkarahisar, 03030, Türkiye
| | - Sadik Kaga
- Department of Biomedical Engineering, Afyon Kocatepe University, Afyonkarahisar, 03200, Türkiye
| | - Korhan Altunbas
- Department of Histology and Embryology, Afyon Kocatepe University, Afyonkarahisar, 03200, Türkiye
| | - Nurullah Okumus
- Department of Pediatrics, Afyonkarahisar Health Sciences University, Afyonkarahisar, 03030, Türkiye
| |
Collapse
|
4
|
Kumar D, Kanchan R, Chaturvedi NK. Targeting protein synthesis pathways in MYC-amplified medulloblastoma. Discov Oncol 2025; 16:23. [PMID: 39779613 PMCID: PMC11711608 DOI: 10.1007/s12672-025-01761-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
MYC is one of the most deregulated oncogenic transcription factors in human cancers. MYC amplification/or overexpression is most common in Group 3 medulloblastoma and is positively associated with poor prognosis. MYC is known to regulate the transcription of major components of protein synthesis (translation) machinery, leading to promoted rates of protein synthesis and tumorigenesis. MTOR signaling-driven deregulated protein synthesis is widespread in various cancers, including medulloblastoma, which can promote the stabilization of MYC. Indeed, our previous studies demonstrate that the key components of protein synthesis machinery, including mTOR signaling and MYC targets, are overexpressed and activated in MYC-amplified medulloblastoma, confirming MYC-dependent addiction of enhanced protein synthesis in medulloblastoma. Further, targeting this enhanced protein synthesis pathway with combined inhibition of MYC transcription and mTOR translation by small-molecule inhibitors, demonstrates preclinical synergistic anti-tumor potential against MYC-driven medulloblastoma in vitro and in vivo. Thus, inhibiting enhanced protein synthesis by targeting the MYC indirectly and mTOR pathways together may present a highly appropriate strategy for treating MYC-driven medulloblastoma and other MYC-addicted cancers. Evidence strongly proposes that MYC/mTOR-driven tumorigenic signaling can predominantly control the translational machinery to elicit cooperative effects on increased cell proliferation, cell cycle progression, and genome dysregulation as a mechanism of cancer initiation. Several small molecule inhibitors of targeting MYC indirectly and mTOR signaling have been developed and used clinically with immunosuppressants and chemotherapy in multiple cancers. Only a few of them have been investigated as treatments for medulloblastoma and other pediatric tumors. This review explores concurrent targeting of MYC and mTOR signaling against MYC-driven medulloblastoma. Based on existing evidence, targeting of MYC and mTOR pathways together produces functional synergy that could be the basis for effective therapies against medulloblastoma.
Collapse
Affiliation(s)
- Devendra Kumar
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 986395, USA
| | - Ranjana Kanchan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nagendra K Chaturvedi
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 986395, USA.
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
5
|
Cox AD, Der CJ. "Undruggable KRAS": druggable after all. Genes Dev 2025; 39:132-162. [PMID: 39638567 PMCID: PMC11789494 DOI: 10.1101/gad.352081.124] [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] [Indexed: 12/07/2024]
Abstract
The three RAS genes (HRAS, KRAS, and NRAS) comprise the most frequently mutated oncogene family in cancer. KRAS is the predominant isoform mutated in cancer and is most prevalently mutated in major causes of cancer deaths including lung, colorectal, and pancreatic cancers. Despite extensive academic and industry efforts to target KRAS, it would take nearly four decades before approval of the first clinically effective KRAS inhibitors for the treatment of KRAS mutant lung cancer. We revisit past anti-KRAS strategies and painful lessons learned and then focus on the rapidly evolving landscape of direct RAS inhibitors, resistance mechanisms, and potential combination treatments.
Collapse
Affiliation(s)
- Adrienne D Cox
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Channing J Der
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
6
|
Slly AM, Ewes WA, Bayoumi WA, Selim KB. Unveiling the potential anticancer activity of new dihydropyrimidines through dual inhibition of EGFR and TrkA: Design, synthesis, and in silico study. Bioorg Chem 2025; 154:107962. [PMID: 39591690 DOI: 10.1016/j.bioorg.2024.107962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024]
Abstract
A series of designed scaffold of dihydropyrimidine was synthesized as dual tyrosine kinase targets inhibitors using a multicomponent Biginelli reaction which provided a high atom economy in a single pot reaction. Several 1,4-DHPM hybrids were obtained via alkylation with different chloroacetylamine derivatives. All the synthesized derivatives were screened for their antiproliferative efficacy towards various cancer cell lines (HCT-116, PC-3, and MCF-7) and normal cell line WI-38 using MTT assay. The results indicated that compounds 8h and 8i have the most significant inhibitory effect on all evaluated cancer cell lines, displaying IC50 of 3.94-15.78 µM. Also, they demonstrated favorable selectivity towards normal cell lines. Moreover, the most active hybrids 8h and 8i were evaluated for their EGFR and TrkA inhibitory activity. The findings indicated that compound 8h had superior inhibitory activity compared to compound 8i on the targeted kinases, effectively stopping the G1 phase of the MCF-7 cell cycle and encouraging apoptosis. Additionally, the molecular docking studies declared that the most active compounds exhibited a notable binding interaction with the binding site of the target proteins. Furthermore, their physicochemical properties, ADMET profiles, and bioavailability radar plots were predicted and analyzed.
Collapse
Affiliation(s)
- Aya M Slly
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Wafaa A Ewes
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Waleed A Bayoumi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Khalid B Selim
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
7
|
Ahmadi Daryakenari N, Wang S, Karniadakis G. CMINNs: Compartment model informed neural networks - Unlocking drug dynamics. Comput Biol Med 2025; 184:109392. [PMID: 39608036 DOI: 10.1016/j.compbiomed.2024.109392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/21/2024] [Accepted: 11/07/2024] [Indexed: 11/30/2024]
Abstract
In the field of pharmacokinetics and pharmacodynamics (PKPD) modeling, which plays a pivotal role in the drug development process, traditional models frequently encounter difficulties in fully encapsulating the complexities of drug absorption, distribution, and their impact on targets. Although multi-compartment models are frequently utilized to elucidate intricate drug dynamics, they can also be overly complex. To generalize modeling while maintaining simplicity, we propose an innovative approach that enhances PK and integrated PK-PD modeling by incorporating fractional calculus or time-varying parameter(s), combined with constant or piecewise constant parameters. These approaches effectively model anomalous diffusion, thereby capturing drug trapping and escape rates in heterogeneous tissues, which is a prevalent phenomenon in drug dynamics. Furthermore, this method provides insight into the dynamics of drug in cancer in multi-dose administrations. Our methodology employs a Physics-Informed Neural Network (PINN) and fractional Physics-Informed Neural Networks (fPINNs), integrating ordinary differential equations (ODEs) with integer/fractional derivative order from compartmental modeling with neural networks. This integration optimizes parameter estimation for variables that are time-variant, constant, piecewise constant, or related to the fractional derivative order. The results demonstrate that this methodology offers a robust framework that not only markedly enhances the model's depiction of drug absorption rates and distributed delayed responses but also unlocks different drug-effect dynamics, providing new insights into absorption rates, anomalous diffusion, drug resistance, persistence, and pharmacokinetic tolerance, all within a system of just two (fractional) ODEs with explainable results. These findings have the potential to streamline drug development by improving the prediction of drug behavior in complex biological systems and shedding light on cancer cell death mechanisms, ultimately aiding in the design of more effective therapeutic strategies.
Collapse
Affiliation(s)
| | - Shupeng Wang
- Division of Applied Mathematics, Brown University, Providence, RI, USA
| | | |
Collapse
|
8
|
Ghosh S, Sharma A, Kumar RS, Nasare V. Sorcin: mechanisms of action in cancer hallmarks, drug resistance and opportunities in therapeutics. Med Oncol 2024; 42:29. [PMID: 39673665 DOI: 10.1007/s12032-024-02580-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Soluble resistant related calcium binding protein (Sorcin) plays an important role in tumor progression, angiogenesis, metastasis, and multidrug resistance. Differential expression of Sorcin across different cancers significantly correlates with key clinicopathological characteristics and survival outcomes, underscoring its potential as a prognostic marker. Its involvement in drug-resistant cancers further advert Sorcin as a promising therapeutic target. This review summarizes the mechanistic role of Sorcin in cancer, its contribution to drug resistance, clinical relevance, and the current and emerging therapeutic approaches aimed at translating Sorcin-targeted therapies into clinical practice.
Collapse
Affiliation(s)
- Sushmita Ghosh
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, Kolkata, India
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, India
| | - Arpana Sharma
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, Kolkata, India
| | - R Suresh Kumar
- Molecular Biology Division, National Institute of Cancer Prevention and Research, ICMR, Noida, Delhi, India
| | - Vilas Nasare
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, Kolkata, India.
| |
Collapse
|
9
|
Malier M, Laverriere MH, Henry M, Yakoubi M, Bellaud P, Arellano C, Sébillot A, Thomas F, Josserand V, Girard E, Roth GS, Millet A. Tumor-associated macrophages confer resistance to chemotherapy (Trifluridine/Tipiracil) in digestive cancers by overexpressing thymidine phosphorylase. Cancer Lett 2024; 606:217307. [PMID: 39454852 DOI: 10.1016/j.canlet.2024.217307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
Pyrimidine analogs are part of the first-line chemotherapy regimen for gastrointestinal cancers. Trifluridine combined with tipiracil, a specific thymidine phosphorylase inhibitor, in TAS-102 has recently emerged as a potential alternative in the face of primary or secondary chemoresistance to 5-fluorouracil. Despite its promise, we report that macrophage-specific overexpression of thymidine phosphorylase results in macrophage-induced chemoresistance to TAS-102 that is insensitive to tipiracil inhibition. Furthermore, we illustrate the human-specific nature of this mechanism, as mouse macrophages do not express substantial levels of thymidine phosphorylase, which constrains the applicability of mouse models. To study the importance of macrophages in chemoresistance to trifluridine, we developed a humanized mouse model with tumor-implanted human macrophages and demonstrated their important role in treatment resistance to pyrimidine analogs. Additionally, our findings revealed that macrophages represent a significant source of thymidine phosphorylase expression, comprising over 40 % of the expressing cells, in human colorectal cancer, thereby contributing to chemoresistance.
Collapse
Affiliation(s)
- Marie Malier
- Univ.Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Team Mechanobiology, Immunity and Cancer, 38000, Grenoble, France
| | - Marie-Hélène Laverriere
- Univ.Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Team Mechanobiology, Immunity and Cancer, 38000, Grenoble, France; Department of Pathology, DACP, University Hospital, Grenoble, France
| | - Maxime Henry
- Univ.Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences OPTIMAL platform, Grenoble, France
| | - Malika Yakoubi
- CRCT Inserm U037, Toulouse University 3, Toulouse, France
| | - Pascale Bellaud
- Univ Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, France-BioImaging (ANR-10-INBS-04), Core Facility H2P2, F-35000, Rennes, France
| | | | - Anthony Sébillot
- Univ Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, France-BioImaging (ANR-10-INBS-04), Core Facility H2P2, F-35000, Rennes, France
| | | | - Véronique Josserand
- Univ.Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences OPTIMAL platform, Grenoble, France
| | - Edouard Girard
- Univ.Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000, Grenoble, France
| | - Gael S Roth
- Univ.Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Team Mechanobiology, Immunity and Cancer, 38000, Grenoble, France; Hepato-Gastroenterology and Digestive Oncology department, University Hospital, Grenoble, France
| | - Arnaud Millet
- Univ.Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Team Mechanobiology, Immunity and Cancer, 38000, Grenoble, France; Hepato-Gastroenterology and Digestive Oncology department, University Hospital, Grenoble, France.
| |
Collapse
|
10
|
Gupta M, Choi H, Kemp SB, Furth EE, Pickup S, Clendenin C, Orlen M, Rosen M, Liu F, Cao Q, Stanger BZ, Zhou R. Quantitative MRI Measurements Capture Pancreatic Cancer and Stroma Reactions to New KRAS Inhibitor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624844. [PMID: 39651222 PMCID: PMC11623539 DOI: 10.1101/2024.11.22.624844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
In pancreatic ductal adenocarcinoma (PDAC), KRAS mutations drive both cancer cell growth and formation of a dense stroma. Small molecule KRAS inhibitors (KRASi) represent a breakthrough for PDAC treatment hence clinical tools that can assess early response, detect resistance and/or predict prolonged survival are desirable for management of patients undergoing KRASi therapy. We hypothesized that diffusion-weighted MRI (DWI) can detect cell death while dynamic contrast enhanced MRI (DCE) and magnetization transfer ratio (MTR) imaging are sensitive to tumor microenvironment changes, and these metrics shed insights into tumor size (standard care assessment) change induced by KRASi treatment. We tested this hypothesis in multiple preclinical PDAC models receiving MRTX1133, an investigational new drug specific for KRAS G12D mutation. Quantitative imaging markers corroborated by immunohistochemistry (IHC) revealed significant and profound changes related to cell death accompanied by changes in tumor cellularity, capillary perfusion /permeability and stromal matrix as early as 48h and day-7 after initiation of KRASi treatment, and greatly prolonged median survival over controls in a genetic engineered mouse model of PDAC (KPC). The MRI markers also captured distinct responses to KRASi therapy from PDAC tumors carrying KRAS G12C versus KRAS G12D mutation. In tumors developed resistance to MRTX1133, the imaging markers exhibited a reversal from those of responding tumors. Our findings have established that multiparametric MRI provide biological insights including cell death, reduced cellularity and tumor microenvironment changes induced by KRASi treatment and set the stage for testing the utility of these clinically ready MRI methods in patients receiving KRASi therapy. Translational relevance Emerging small molecule KRAS inhibitors (KRASi) represent a new class of therapy for PDAC. Clinical tools that can provide biological insights beyond tumor size change are desirable for management of patients under KRASi therapy. DWI and DCE are frequently applied MRI methods for assessing cancer treatment responses in clinical trials. Using multiple PDAC models, we examined whether DWI, DCE and MTR can enhance the standard care assessment (tumor size) to MRTX1133, a KARSi with investigational new drug (IND) status. Our data demonstrate the abilities of DWI, DCE and MTR derived imaging markers to detect the early (48h) cell death, pronounced stromal changes and development of resistance to KRASi. This study has high translational relevance by testing clinically ready MRI methods, an IND and a genetic engineered mouse model that recapitulates saline features of human PDAC.
Collapse
|
11
|
Connell W, Garcia K, Goodarzi H, Keiser MJ. Learning chemical sensitivity reveals mechanisms of cellular response. Commun Biol 2024; 7:1149. [PMID: 39278951 PMCID: PMC11402971 DOI: 10.1038/s42003-024-06865-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 09/06/2024] [Indexed: 09/18/2024] Open
Abstract
Chemical probes interrogate disease mechanisms at the molecular level by linking genetic changes to observable traits. However, comprehensive chemical screens in diverse biological models are impractical. To address this challenge, we develop ChemProbe, a model that predicts cellular sensitivity to hundreds of molecular probes and drugs by learning to combine transcriptomes and chemical structures. Using ChemProbe, we infer the chemical sensitivity of cancer cell lines and tumor samples and analyze how the model makes predictions. We retrospectively evaluate drug response predictions for precision breast cancer treatment and prospectively validate chemical sensitivity predictions in new cellular models, including a genetically modified cell line. Our model interpretation analysis identifies transcriptome features reflecting compound targets and protein network modules, identifying genes that drive ferroptosis. ChemProbe is an interpretable in silico screening tool that allows researchers to measure cellular response to diverse compounds, facilitating research into molecular mechanisms of chemical sensitivity.
Collapse
Affiliation(s)
- William Connell
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Kristle Garcia
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Hani Goodarzi
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Michael J Keiser
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA.
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
12
|
Rajaram J, Mende LK, Kuthati Y. A Review of the Efficacy of Nanomaterial-Based Natural Photosensitizers to Overcome Multidrug Resistance in Cancer. Pharmaceutics 2024; 16:1120. [PMID: 39339158 PMCID: PMC11434998 DOI: 10.3390/pharmaceutics16091120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/27/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Natural photosensitizers (PS) are compounds derived from nature, with photodynamic properties. Natural PSs have a similar action to that of commercial PSs, where cancer cell death occurs by necrosis, apoptosis, and autophagy through ROS generation. Natural PSs have garnered great interest over the last few decades because of their high biocompatibility and good photoactivity. Specific wavelengths could cause phytochemicals to produce harmful ROS for photodynamic therapy (PDT). However, natural PSs have some shortcomings, such as reduced solubility and lower uptake, making them less appropriate for PDT. Nanotechnology offers an opportunity to develop suitable carriers for various natural PSs for PDT applications. Various nanoparticles have been developed to improve the outcome with enhanced solubility, optical adsorption, and tumor targeting. Multidrug resistance (MDR) is a phenomenon in which tumor cells develop resistance to a wide range of structurally and functionally unrelated drugs. Over the last decade, several researchers have extensively studied the effect of natural PS-based photodynamic treatment (PDT) on MDR cells. Though the outcomes of clinical trials for natural PSs were inconclusive, significant advancement is still required before PSs can be used as a PDT agent for treating MDR tumors. This review addresses the increasing literature on MDR tumor progression and the efficacy of PDT, emphasizing the importance of developing new nano-based natural PSs in the fight against MDR that have the required features for an MDR tumor photosensitizing regimen.
Collapse
Affiliation(s)
- Jagadeesh Rajaram
- Department of Biochemistry and Molecular Medicine, National Dong Hwa University, Hualien 974, Taiwan;
| | - Lokesh Kumar Mende
- Department of Anesthesiology, Cathy General Hospital, Taipei 106, Taiwan;
| | - Yaswanth Kuthati
- Department of Anesthesiology, Cathy General Hospital, Taipei 106, Taiwan;
| |
Collapse
|
13
|
Moure CJ, Vara B, Cheng MM, Sondey C, Muise E, Park E, Vela Ramirez JE, Su D, D'Souza S, Yan Q, Yeung CS, Zhang M, Mansueto MS, Linn D, Buchanan M, Foti R, DiMauro E, Long B, Simov V, Barry ER. Activation of Hepatocyte Growth Factor/MET Signaling as a Mechanism of Acquired Resistance to a Novel YAP1/TEAD Small Molecule Inhibitor. Mol Cancer Ther 2024; 23:1095-1108. [PMID: 38691847 DOI: 10.1158/1535-7163.mct-23-0538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/21/2023] [Accepted: 03/04/2024] [Indexed: 05/03/2024]
Abstract
Many tumor types harbor alterations in the Hippo pathway, including mesothelioma, where a high percentage of cases are considered YAP1/TEAD dependent. Identification of autopalmitoylation sites in the hydrophobic palmitate pocket of TEADs, which may be necessary for YAP1 protein interactions, has enabled modern drug discovery platforms to generate compounds that allosterically inhibit YAP1/TEAD complex formation and transcriptional activity. We report the discovery and characterization of a novel YAP1/TEAD inhibitor MRK-A from an aryl ether chemical series demonstrating potent and specific inhibition of YAP1/TEAD activity. In vivo, MRK-A showed a favorable tolerability profile in mice and demonstrated pharmacokinetics suitable for twice daily oral dosing in preclinical efficacy studies. Importantly, monotherapeutic targeting of YAP1/TEAD in preclinical models generated regressions in a mesothelioma CDX model; however, rapid resistance to therapy was observed. RNA-sequencing of resistant tumors revealed mRNA expression changes correlated with the resistance state and a marked increase of hepatocyte growth factor (HGF) expression. In vitro, exogenous HGF was able to fully rescue cytostasis induced by MRK-A in mesothelioma cell lines. In addition, co-administration of small molecule inhibitors of the MET receptor tyrosine kinase suppressed the resistance generating effect of HGF on MRK-A induced growth inhibition. In this work, we report the structure and characterization of MRK-A, demonstrating potent and specific inhibition of YAP1/TAZ-TEAD-mediated transcriptional responses, with potential implications for treating malignancies driven by altered Hippo signaling, including factors resulting in acquired drug resistance.
Collapse
Affiliation(s)
- Casey J Moure
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| | - Brandon Vara
- Department of Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey
| | - Mangeng M Cheng
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| | - Christopher Sondey
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| | - Eric Muise
- Department of Data and Genome Sciences, Merck & Co., Inc., Rahway, New Jersey
| | - Eunsil Park
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| | | | - Dan Su
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| | - Shanti D'Souza
- Department of Discovery Oncology, Merck & Co., Inc., Rahway, New Jersey
| | - Qingyun Yan
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| | - Charles S Yeung
- Department of Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey
| | - Minjia Zhang
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| | - My Sam Mansueto
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| | - Doug Linn
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| | - Mark Buchanan
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| | - Robert Foti
- Department of PPDM, Merck & Co., Inc., Rahway, New Jersey
| | - Erin DiMauro
- Department of Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey
| | - Brian Long
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| | - Vladimir Simov
- Department of Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey
| | - Evan R Barry
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey
| |
Collapse
|
14
|
Garg P, Malhotra J, Kulkarni P, Horne D, Salgia R, Singhal SS. Emerging Therapeutic Strategies to Overcome Drug Resistance in Cancer Cells. Cancers (Basel) 2024; 16:2478. [PMID: 39001539 PMCID: PMC11240358 DOI: 10.3390/cancers16132478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
The rise of drug resistance in cancer cells presents a formidable challenge in modern oncology, necessitating the exploration of innovative therapeutic strategies. This review investigates the latest advancements in overcoming drug resistance mechanisms employed by cancer cells, focusing on emerging therapeutic modalities. The intricate molecular insights into drug resistance, including genetic mutations, efflux pumps, altered signaling pathways, and microenvironmental influences, are discussed. Furthermore, the promising avenues offered by targeted therapies, combination treatments, immunotherapies, and precision medicine approaches are highlighted. Specifically, the synergistic effects of combining traditional cytotoxic agents with molecularly targeted inhibitors to circumvent resistance pathways are examined. Additionally, the evolving landscape of immunotherapeutic interventions, including immune checkpoint inhibitors and adoptive cell therapies, is explored in terms of bolstering anti-tumor immune responses and overcoming immune evasion mechanisms. Moreover, the significance of biomarker-driven strategies for predicting and monitoring treatment responses is underscored, thereby optimizing therapeutic outcomes. For insights into the future direction of cancer treatment paradigms, the current review focused on prevailing drug resistance challenges and improving patient outcomes, through an integrative analysis of these emerging therapeutic strategies.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, India
| | - Jyoti Malhotra
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
15
|
Chis AA, Dobrea CM, Arseniu AM, Frum A, Rus LL, Cormos G, Georgescu C, Morgovan C, Butuca A, Gligor FG, Vonica-Tincu AL. Antibody-Drug Conjugates-Evolution and Perspectives. Int J Mol Sci 2024; 25:6969. [PMID: 39000079 PMCID: PMC11241239 DOI: 10.3390/ijms25136969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Antineoplastic therapy is one of the main research themes of this century. Modern approaches have been implemented to target and heighten the effect of cytostatic drugs on tumors and diminish their general/unspecific toxicity. In this context, antibody-drug conjugates (ADCs) represent a promising and successful strategy. The aim of this review was to assess different aspects regarding ADCs. They were presented from a chemical and a pharmacological perspective and aspects like structure, conjugation and development particularities alongside effects, clinical trials, safety issues and perspectives and challenges for future use of these drugs were discussed. Representative examples include but are not limited to the following main structural components of ADCs: monoclonal antibodies (trastuzumab, brentuximab), linkers (pH-sensitive, reduction-sensitive, peptide-based, phosphate-based, and others), and payloads (doxorubicin, emtansine, ravtansine, calicheamicin). Regarding pharmacotherapy success, the high effectiveness expectation associated with ADC treatment is supported by the large number of ongoing clinical trials. Major aspects such as development strategies are first discussed, advantages and disadvantages, safety and efficacy, offering a retrospective insight on the subject. The second part of the review is prospective, focusing on various plans to overcome the previously identified difficulties.
Collapse
Affiliation(s)
| | | | - Anca Maria Arseniu
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Adina Frum
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Luca-Liviu Rus
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Gabriela Cormos
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Cecilia Georgescu
- Faculty of Agriculture Science, Food Industry and Environmental Protection, "Lucian Blaga" University of Sibiu, 550012 Sibiu, Romania
| | - Claudiu Morgovan
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Anca Butuca
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | | | | |
Collapse
|
16
|
Meng Q, Han J, Wang P, Jia C, Guan M, Zhang B, Zhao W. BMS-794833 reduces anlotinib resistance in osteosarcoma by targeting the VEGFR/Ras/CDK2 pathway. J Bone Oncol 2024; 45:100594. [PMID: 38532893 PMCID: PMC10963651 DOI: 10.1016/j.jbo.2024.100594] [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: 11/21/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Background Osteosarcoma, a tumor that originates from bone cells, has a poor prognosis and a high degree of malignancy. Anlotinib, a small-molecule multi-target tyrosine kinase inhibitor (TKI), is the first-line drug in treating osteosarcoma, especially in late-stage osteosarcoma. However, patients often develop resistance after using anlotinib for a certain period, which poses a challenge to its further clinical application. Recently, several TKIs, for instance regorafenib and cabozantinib, have showed clinical interest in treating osteosarcoma and target both vascular endothelial growth factor receptor (VEGFR) and mesenchymal epithelial transition factor (c-MET). Therefore, the identification of new TKI warrants further investigation. Methods We performed CCK8 aasays to confirm that BMS-794833 sensitization osteosarcoma cells to anlotinib. Bioinformatics analysis and rescue experiments showed that the reduce of resistance were dependent on the VEGFR/Ras/CDK2 pathway. Cell line based xenograft model were used to demonstrate that BMS-794833 and anlotinib could synergistically treat OS. Results Here, we found that BMS-794833 reduced anlotinib resistance in osteosarcoma by targeting the VEGFR/Ras/CDK2 pathway. CCK8 assay showed that BMS-794833 significantly improved the resistance of osteosarcoma cells to anlotinib. The results of rescue experiments showed that the regulatory effects of BMS-794833 on the proliferation and drug resistance of osteosarcoma cells were dependent on the VEGFR/Ras/CDK2 pathway. In addition, BMS-794833 affected the resistance of osteosarcoma cells to anlotinib through epithelial-mesenchymal transition (EMT) and apoptosis pathways. More importantly, BMS-794833 and anlotinib exerted synergistic therapeutic effects against osteosarcoma in vivo. Conclusion Altogether, this study reveals a new (VEGFR)-targeting drug that can be combined with anlotinib for the treatment of osteosarcoma, which provides an important theoretical basis for overcoming anlotinib resistance.
Collapse
Affiliation(s)
- Qingtao Meng
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, Dalian 116028, China
- Department of Orthopedics, Dalian NO.3 People’s Hospital, Dalian 116091, China
| | - Jian Han
- Department of Orthopedics, Dalian NO.3 People’s Hospital, Dalian 116091, China
| | - Peng Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Chenxu Jia
- Department of Orthopedics, Dalian NO.3 People’s Hospital, Dalian 116091, China
| | - Mingyang Guan
- Department of Orthopedics, Dalian NO.3 People’s Hospital, Dalian 116091, China
| | - Bolun Zhang
- Department of Orthopedics, Dalian NO.3 People’s Hospital, Dalian 116091, China
| | - Wenzhi Zhao
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, Dalian 116028, China
| |
Collapse
|
17
|
Taverna JA, Hung CN, Williams M, Williams R, Chen M, Kamali S, Sambandam V, Hsiang-Ling Chiu C, Osmulski PA, Gaczynska ME, DeArmond DT, Gaspard C, Mancini M, Kusi M, Pandya AN, Song L, Jin L, Schiavini P, Chen CL. Ex vivo drug testing of patient-derived lung organoids to predict treatment responses for personalized medicine. Lung Cancer 2024; 190:107533. [PMID: 38520909 DOI: 10.1016/j.lungcan.2024.107533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/25/2024]
Abstract
Lung cancer is the leading cause of global cancer-related mortality resulting in ∼ 1.8 million deaths annually. Systemic, molecular targeted, and immune therapies have provided significant improvements of survival outcomes for patients. However, drug resistance usually arises and there is an urgent need for novel therapy screening and personalized medicine. 3D patient-derived organoid (PDO) models have emerged as a more effective and efficient alternative for ex vivo drug screening than 2D cell culture and patient-derived xenograft (PDX) models. In this review, we performed an extensive search of lung cancer PDO-based ex vivo drug screening studies. Lung cancer PDOs were successfully established from fresh or bio-banked sections and/or biopsies, pleural effusions and PDX mouse models. PDOs were subject to ex vivo drug screening with chemotherapy, targeted therapy and/or immunotherapy. PDOs consistently recapitulated the genomic alterations and drug sensitivity of primary tumors. Although sample sizes of the previous studies were limited and some technical challenges remain, PDOs showed great promise in the screening of novel therapy drugs. With the technical advances of high throughput, tumor-on-chip, and combined microenvironment, the drug screening process using PDOs will enhance precision care of lung cancer patients.
Collapse
Affiliation(s)
- Josephine A Taverna
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA; Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; Department of Medicine, Division of Hematology and Oncology, University of Texas Health Science Center, San Antonio, TX, USA.
| | - Chia-Nung Hung
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Madison Williams
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; Department of Medicine, Division of Hematology and Oncology, University of Texas Health Science Center, San Antonio, TX, USA; Department of General Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ryan Williams
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; Department of Medicine, Division of Hematology and Oncology, University of Texas Health Science Center, San Antonio, TX, USA; Department of General Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meizhen Chen
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | | | | | - Cheryl Hsiang-Ling Chiu
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Pawel A Osmulski
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Maria E Gaczynska
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Daniel T DeArmond
- Department of Medicine, Division of Hematology and Oncology, University of Texas Health Science Center, San Antonio, TX, USA; Department of General Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, Texas and Department of Laboratory Medicine, Baptist Health System, San Antonio, TX, USA
| | - Christine Gaspard
- Dolph Briscoe, Jr. Library, University of Texas Health Science Center, San Antonio, TX, USA
| | | | - Meena Kusi
- Deciphera Pharmaceuticals, LLC., Waltham, MA, USA
| | - Abhishek N Pandya
- Department of Medicine, Division of Hematology and Oncology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Lina Song
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Lingtao Jin
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | | | - Chun-Liang Chen
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA; Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; School of Nursing, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
18
|
Lamichhane A, Tavana H. Three-Dimensional Tumor Models to Study Cancer Stemness-Mediated Drug Resistance. Cell Mol Bioeng 2024; 17:107-119. [PMID: 38737455 PMCID: PMC11082110 DOI: 10.1007/s12195-024-00798-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/01/2024] [Indexed: 05/14/2024] Open
Abstract
Solid tumors often contain genetically different populations of cancer cells, stromal cells, various structural and soluble proteins, and other soluble signaling molecules. The American Cancer society estimated 1,958,310 new cancer cases and 609,820 cancer deaths in the United States in 2023. A major barrier against successful treatment of cancer patients is drug resistance. Gain of stem cell-like states by cancer cells under drug pressure or due to interactions with the tumor microenvironment is a major mechanism that renders therapies ineffective. Identifying approaches to target cancer stem cells is expected to improve treatment outcomes for patients. Most of our understanding of drug resistance and the role of cancer stemness is from monolayer cell cultures. Recent advances in cell culture technologies have enabled developing sophisticated three-dimensional tumor models that facilitate mechanistic studies of cancer drug resistance. This review summarizes the role of cancer stemness in drug resistance and highlights the various tumor models that are used to discover the underlying mechanisms and test potentially novel therapeutics.
Collapse
Affiliation(s)
- Astha Lamichhane
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325 USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325 USA
| |
Collapse
|
19
|
Chen M, Mainardi S, Lieftink C, Velds A, de Rink I, Yang C, Kuiken HJ, Morris B, Edwards F, Jochems F, van Tellingen O, Boeije M, Proost N, Jansen RA, Qin S, Jin H, Koen van der Mijn JC, Schepers A, Venkatesan S, Qin W, Beijersbergen RL, Wang L, Bernards R. Targeting of vulnerabilities of drug-tolerant persisters identified through functional genetics delays tumor relapse. Cell Rep Med 2024; 5:101471. [PMID: 38508142 PMCID: PMC10983104 DOI: 10.1016/j.xcrm.2024.101471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 12/01/2023] [Accepted: 02/21/2024] [Indexed: 03/22/2024]
Abstract
Drug-tolerant persisters (DTPs) are a rare subpopulation of cells within a tumor that can survive therapy through nongenetic adaptive mechanisms to develop relapse and repopulate the tumor following drug withdrawal. Using a cancer cell line with an engineered suicide switch to kill proliferating cells, we perform both genetic screens and compound screens to identify the inhibition of bromodomain and extraterminal domain (BET) proteins as a selective vulnerability of DTPs. BET inhibitors are especially detrimental to DTPs that have reentered the cell cycle (DTEPs) in a broad spectrum of cancer types. Mechanistically, BET inhibition induces lethal levels of ROS through the suppression of redox-regulating genes highly expressed in DTPs, including GPX2, ALDH3A1, and MGST1. In vivo BET inhibitor treatment delays tumor relapse in both melanoma and lung cancer. Our study suggests that combining standard of care therapy with BET inhibitors to eliminate residual persister cells is a promising therapeutic strategy.
Collapse
Affiliation(s)
- Mengnuo Chen
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sara Mainardi
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Cor Lieftink
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands; NKI Robotics and Screening Center, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Arno Velds
- Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Iris de Rink
- Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Chen Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hendrik J Kuiken
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands; NKI Robotics and Screening Center, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ben Morris
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands; NKI Robotics and Screening Center, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Finn Edwards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Fleur Jochems
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Manon Boeije
- Mouse Clinic for Cancer and Aging Research, Preclinical Intervention Unit, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Natalie Proost
- Mouse Clinic for Cancer and Aging Research, Preclinical Intervention Unit, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Robin A Jansen
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Shifan Qin
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Haojie Jin
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - J C Koen van der Mijn
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Arnout Schepers
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Subramanian Venkatesan
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Roderick L Beijersbergen
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands; NKI Robotics and Screening Center, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
20
|
McDonald PC, Dedhar S. Persister cell plasticity in tumour drug resistance. Semin Cell Dev Biol 2024; 156:1-10. [PMID: 37977107 DOI: 10.1016/j.semcdb.2023.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
The emergence of therapeutic resistance remains a formidable barrier to durable responses by cancer patients and is a major cause of cancer-related deaths. It is increasingly recognized that non-genetic mechanisms of acquired resistance are important in many cancers. These mechanisms of resistance rely on inherent cellular plasticity where cancer cells can switch between multiple phenotypic states without genetic alterations, providing a dynamic, reversible resistance landscape. Such mechanisms underlie the generation of drug-tolerant persister (DTP) cells, a subpopulation of tumour cells that contributes to heterogeneity within tumours and that supports therapeutic resistance. In this review, we provide an overview of the major features of DTP cells, focusing on phenotypic and metabolic plasticity as two key drivers of tolerance and persistence. We discuss the link between DTP cell plasticity and the potential vulnerability of these cells to ferroptosis. We also discuss the relationship between DTP cells and cells that survive the induction of apoptosis, a process termed anastasis, and discuss the properties of such cells in the context of increased metastatic potential and sensitivity to cell death mechanisms such as ferroptosis.
Collapse
Affiliation(s)
- Paul C McDonald
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Shoukat Dedhar
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
21
|
Raut KK, Pandey S, Kharel G, Pascal SM. Evidence of direct interaction between cisplatin and the caspase-cleaved prostate apoptosis response-4 tumor suppressor. Protein Sci 2024; 33:e4867. [PMID: 38093605 PMCID: PMC10868438 DOI: 10.1002/pro.4867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/09/2023] [Accepted: 12/11/2023] [Indexed: 02/16/2024]
Abstract
Prostate apoptosis response-4 (Par-4) tumor suppressor protein has gained attention as a potential therapeutic target owing to its unique ability to selectively induce apoptosis in cancer cells, sensitize them to chemotherapy and radiotherapy, and mitigate drug resistance. It has recently been reported that Par-4 interacts synergistically with cisplatin, a widely used anticancer drug. However, the mechanistic details underlying this relationship remain elusive. In this investigation, we employed an array of biophysical techniques, including circular dichroism spectroscopy, dynamic light scattering, and UV-vis absorption spectroscopy, to characterize the interaction between the active caspase-cleaved Par-4 (cl-Par-4) fragment and cisplatin. Additionally, elemental analysis was conducted to quantitatively assess the binding of cisplatin to the protein, utilizing inductively coupled plasma-optical emission spectroscopy and atomic absorption spectroscopy. Our findings provide evidence of direct interaction between cl-Par-4 and cisplatin, and reveal a binding stoichiometry of 1:1. This result provides insights that could be useful in enhancing the efficacy of cisplatin-based and tumor suppressor-based cancer therapies.
Collapse
Affiliation(s)
- Krishna K. Raut
- Department of Chemistry and BiochemistryOld Dominion UniversityNorfolkVirginiaUSA
| | - Samjhana Pandey
- Biomedical Sciences ProgramOld Dominion UniversityNorfolkVirginiaUSA
| | - Gyanendra Kharel
- Department of Chemistry and BiochemistryOld Dominion UniversityNorfolkVirginiaUSA
| | - Steven M. Pascal
- Department of Chemistry and BiochemistryOld Dominion UniversityNorfolkVirginiaUSA
| |
Collapse
|
22
|
Uehara M, Domoto T, Takenaka S, Takeuchi O, Shimasaki T, Miyashita T, Minamoto T. Glycogen synthase kinase 3β: the nexus of chemoresistance, invasive capacity, and cancer stemness in pancreatic cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:4. [PMID: 38318525 PMCID: PMC10838383 DOI: 10.20517/cdr.2023.84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/20/2023] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
The treatment of pancreatic cancer remains a significant clinical challenge due to the limited number of patients eligible for curative (R0) surgery, failures in the clinical development of targeted and immune therapies, and the pervasive acquisition of chemotherapeutic resistance. Refractory pancreatic cancer is typified by high invasiveness and resistance to therapy, with both attributes related to tumor cell stemness. These malignant characteristics mutually enhance each other, leading to rapid cancer progression. Over the past two decades, numerous studies have produced evidence of the pivotal role of glycogen synthase kinase (GSK)3β in the progression of over 25 different cancer types, including pancreatic cancer. In this review, we synthesize the current knowledge on the pathological roles of aberrant GSK3β in supporting tumor cell proliferation and invasion, as well as its contribution to gemcitabine resistance in pancreatic cancer. Importantly, we discuss the central role of GSK3β as a molecular hub that mechanistically connects chemoresistance, tumor cell invasion, and stemness in pancreatic cancer. We also discuss the involvement of GSK3β in the formation of desmoplastic tumor stroma and in promoting anti-cancer immune evasion, both of which constitute major obstacles to successful cancer treatment. Overall, GSK3β has characteristics of a promising therapeutic target to overcome chemoresistance in pancreatic cancer.
Collapse
Affiliation(s)
- Masahiro Uehara
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Authors contributed equally
| | - Takahiro Domoto
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Authors contributed equally
| | - Satoshi Takenaka
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
- Department of Surgery, Toyama City Hospital, Toyama 939-8511, Japan
| | - Osamu Takeuchi
- Biomedical Laboratory, Department of Research, Kitasato University Kitasato Institute Hospital, Tokyo 108-8642, Japan
| | - Takeo Shimasaki
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Japan
| | - Tomoharu Miyashita
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
- Department of Surgery, Toyama City Hospital, Toyama 939-8511, Japan
| | - Toshinari Minamoto
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
| |
Collapse
|
23
|
Zamora PO, Altay G, Santamaria U, Dwarshuis N, Donthi H, Moon CI, Bakalar D, Zamora M. Drug Responses in Plexiform Neurofibroma Type I (PNF1) Cell Lines Using High-Throughput Data and Combined Effectiveness and Potency. Cancers (Basel) 2023; 15:5811. [PMID: 38136356 PMCID: PMC10742026 DOI: 10.3390/cancers15245811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Background: Neurofibromatosis type 1 (NF1) is a genetic disorder characterized by heterozygous germline NF1 gene mutations that predispose patients to developing plexiform neurofibromas, which are benign but often disfiguring tumors of the peripheral nerve sheath induced by loss of heterozygosity at the NF1 locus. These can progress to malignant peripheral nerve sheath tumors (MPNSTs). There are no approved drug treatments for adults with NF1-related inoperable plexiform neurofibromas, and only one drug (selumetinib), which is an FDA-approved targeted therapy for the treatment of symptomatic pediatric plexiform neurofibromas, highlighting the need for additional drug screening and development. In high-throughput screening, the effectiveness of drugs against cell lines is often assessed by measuring in vitro potency (AC50) or the area under the curve (AUC). However, the variability of dose-response curves across drugs and cell lines and the frequency of partial effectiveness suggest that these measures alone fail to provide a full picture of overall efficacy. Methods: Using concentration-response data, we combined response effectiveness (EFF) and potency (AC50) into (a) a score characterizing the effect of a compound on a single cell line, S = log[EFF/AC50], and (b) a relative score, ΔS, characterizing the relative difference between a reference (e.g., non-tumor) and test (tumor) cell line. ΔS was applied to data from high-throughput screening (HTS) of a drug panel tested on NF1-/- tumor cells, using immortalized non-tumor NF1+/- cells as a reference. Results: We identified drugs with sensitivity, targeting expected pathways, such as MAPK-ERK and PI3K-AKT, as well as serotonin-related targets, among others. The ΔS technique used here, in tandem with a supplemental ΔS web tool, simplifies HTS analysis and may provide a springboard for further investigations into drug response in NF1-related cancers. The tool may also prove useful for drug development in a variety of other cancers.
Collapse
Affiliation(s)
| | | | | | | | | | - Chang In Moon
- Dan L. Duncan Comprehensive Cancer Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dana Bakalar
- National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
24
|
Wang X, Eichhorn PJA, Thiery JP. TGF-β, EMT, and resistance to anti-cancer treatment. Semin Cancer Biol 2023; 97:1-11. [PMID: 37944215 DOI: 10.1016/j.semcancer.2023.10.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 05/08/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
Transforming growth factor-β (TGF-β) signaling regulates cell-specific programs involved in embryonic development, wound-healing, and immune homeostasis. Yet, during tumor progression, these TGF-β-mediated programs are altered, leading to epithelial cell plasticity and a reprogramming of epithelial cells into mesenchymal lineages through epithelial-to-mesenchymal transition (EMT), a critical developmental program in morphogenesis and organogenesis. These changes, in turn, lead to enhanced carcinoma cell invasion, metastasis, immune cell differentiation, immune evasion, and chemotherapy resistance. Here, we discuss EMT as one of the critical programs associated with carcinoma cell plasticity and the influence exerted by TGF-β on carcinoma status and function. We further explore the composition of carcinoma and other cell populations within the tumor microenvironment, and consider the relevant outcomes related to the programs associated with cancer treatment resistance.
Collapse
Affiliation(s)
- Xuecong Wang
- Guangzhou National Laboratory, Guangzhou, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Pieter Johan Adam Eichhorn
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, GPO Box U1987, Perth, WA 6845, Australia; Curtin Medical School, Curtin University, GPO Box U1987, Perth, WA 6845, Australia; Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore, Singapore
| | | |
Collapse
|
25
|
Zhang Y, Xu H, Pi S, Tan H, Huang B, Chen Y. The prognostic and immunological role of FKBP1A in an integrated muti-omics cancers analysis, especially lung cancer. J Cancer Res Clin Oncol 2023; 149:16589-16608. [PMID: 37715833 DOI: 10.1007/s00432-023-05362-1] [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: 07/19/2023] [Accepted: 08/28/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND AND AIM FKBP1A, a gene encoding the FK506-binding protein 1A, has emerged as a significant player in cancer progression and prognosis. This study aimed to comprehensively investigate the multifaceted role of FKBP1A in cancer, focusing on its differential expression patterns, prognostic implications, genetic alterations, and associations with the tumor microenvironment. METHODS AND RESULTS Using large-scale datasets, including GTEx, TCGA, HPA, and cBioPortal, we analyzed FKBP1A expression across normal tissues and various cancer types. Our findings revealed that FKBP1A exhibited aberrant upregulation in most human cancers, making it a potential biomarker for malignancy. Moreover, FKBP1A expression correlated with poor overall survival, disease-specific survival, disease-free interval, and progression-free interval in several cancers, indicating its prognostic significance. Genetic alteration analysis showed that FKBP1A gene amplification was prevalent, particularly in ovarian cancer. Furthermore, FKBP1A expression was associated with tumor mutational burden and microsatellite instability, highlighting its potential involvement in tumor-immune response. Notably, FKBP1A expression positively correlated with stromal and immune cell scores, suggesting its role in shaping the tumor microenvironment. Additionally, according to the functional enrichment analysis, experimental validation in lung adenocarcinoma confirmed the role of FKBP1A through the regulation of EGFR signaling by apoptosis, which is consistent with drug sensitivity analysis to some extent. CONCLUSION In conclusion, FKBP1A exhibits differential expression in cancer, serves as a prognostic indicator, undergoes genetic alterations, and influences the tumor-immune microenvironment. These findings shed light on the multifaceted role of FKBP1A in cancer development and progression, suggesting its potential as a therapeutic target and guidance of clinical drugs selection, and provide valuable insights into patient prognosis for interventions based on pharmaceuticals.
Collapse
Affiliation(s)
- Yi Zhang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Haifeng Xu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
- Department of Infectious Diseases, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Sainan Pi
- Department of Infectious Diseases, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Huiqian Tan
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
- Department of Infectious Diseases, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Bihui Huang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Youpeng Chen
- Department of Infectious Diseases, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
26
|
McDonald PC, Dedhar S. Co-vulnerabilities of inhibiting carbonic anhydrase IX in ferroptosis-mediated tumor cell death. Front Mol Biosci 2023; 10:1327310. [PMID: 38099193 PMCID: PMC10720035 DOI: 10.3389/fmolb.2023.1327310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023] Open
Abstract
The tumour-associated carbonic anhydrases (CA) IX and XII are upregulated by cancer cells to combat cellular and metabolic stress imparted by hypoxia and acidosis in solid tumours. Owing to its tumour-specific expression and function, CAIX is an attractive therapeutic target and this has driven intense efforts to develop pharmacologic agents to target its activity, including small molecule inhibitors. Many studies in multiple solid tumour models have demonstrated that targeting CAIX activity with the selective CAIX/XII inhibitor, SLC-0111, results in anti-tumour efficacy, particularly when used in combination with chemotherapy or immune checkpoint blockade, and has now advanced to the clinic. However, it has been observed that sustainability and durability of CAIX inhibition, even in combination with chemotherapy agents, is limited by the occurrence of adaptive resistance, resulting in tumour recurrence. Importantly, the data from these models demonstrates that CAIX inhibition may sensitize tumour cells to cytotoxic drugs and evidence now points to ferroptosis, an iron-dependent form of regulated cell death (RCD) that results from accumulation of toxic levels of phospholipid peroxidation as a major mechanism involved in CAIX-mediated sensitization to cancer therapy. In this mini-review, we discuss recent advances demonstrating the mechanistic role CAIX plays in sensitizing cancer cells to ferroptosis.
Collapse
Affiliation(s)
- Paul C. McDonald
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Shoukat Dedhar
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
27
|
Ni Y, Liang Y, Li M, Lin Y, Zou X, Han F, Cao J, Li L. The updates on metastatic mechanism and treatment of colorectal cancer. Pathol Res Pract 2023; 251:154837. [PMID: 37806170 DOI: 10.1016/j.prp.2023.154837] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/20/2023] [Accepted: 09/30/2023] [Indexed: 10/10/2023]
Abstract
Colorectal cancer (CRC) is a main cause of cancer death worldwide. Metastasis is a major cause of cancer-related death in CRC. The treatment of metastatic CRC has progressed minimally. However, the potential molecular mechanisms involved in CRC metastasis have remained to be comprehensively clarified. An improved understanding of the CRC mechanistic determinants is needed to better prevent and treat metastatic cancer. In this review, based on evidence from a growing body of research in metastatic cancers, we discuss the cellular and molecular mechanisms involved in CRC metastasis. This review reveals both the molecular mechanisms of metastases and identifies new opportunities for developing more effective strategies to target metastatic relapse and improve CRC patient outcomes.
Collapse
Affiliation(s)
- Yunfei Ni
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - You Liang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Mingzhou Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Yang Lin
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Xin Zou
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Fangyi Han
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Jianing Cao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Liang Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China.
| |
Collapse
|
28
|
Lamichhane A, Luker GD, Agarwal S, Tavana H. Inhibiting BRAF/EGFR/MEK suppresses cancer stemness and drug resistance of primary colorectal cancer cells. Oncotarget 2023; 14:879-889. [PMID: 37791907 PMCID: PMC10549774 DOI: 10.18632/oncotarget.28517] [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: 05/02/2023] [Accepted: 09/21/2023] [Indexed: 10/05/2023] Open
Abstract
Drug resistance is a major barrier against successful treatments of cancer patients. Gain of stemness under drug pressure is a major mechanism that renders treatments ineffective. Identifying approaches to target cancer stem cells (CSCs) is expected to improve treatment outcomes for patients. To elucidate the role of cancer stemness in resistance of colorectal cancer cells to targeted therapies, we developed spheroid cultures of patient-derived BRAFmut and KRASmut tumor cells and studied resistance mechanisms to inhibition of MAPK pathway through phenotypic and gene and protein expression analysis. We found that treatments enriched the expression of CSC markers CD166, ALDH1A3, CD133, and LGR5 and activated PI3K/Akt pathway in cancer cells. We examined various combination treatments to block these activities and found that a triple combination against BRAF, EGFR, and MEK significantly reduced stemness and activities of oncogenic signaling pathways. This study demonstrates the feasibility of blocking stemness-mediated drug resistance and tumorigenic activities in colorectal cancer.
Collapse
Affiliation(s)
- Astha Lamichhane
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Gary D. Luker
- Department of Radiology, Microbiology and Immunology, Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Seema Agarwal
- Department of Pathology, Biochemistry, Molecular and Cellular Biology, Georgetown University, Washington, DC 20007, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| |
Collapse
|
29
|
Blaquier JB, Ortiz-Cuaran S, Ricciuti B, Mezquita L, Cardona AF, Recondo G. Tackling Osimertinib Resistance in EGFR-Mutant Non-Small Cell Lung Cancer. Clin Cancer Res 2023; 29:3579-3591. [PMID: 37093192 DOI: 10.1158/1078-0432.ccr-22-1912] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/11/2023] [Accepted: 04/12/2023] [Indexed: 04/25/2023]
Abstract
The current landscape of targeted therapies directed against oncogenic driver alterations in non-small cell lung cancer (NSCLC) is expanding. Patients with EGFR-mutant NSCLC can derive significant benefit from EGFR tyrosine kinase inhibitor (TKI) therapy, including the third-generation EGFR TKI osimertinib. However, invariably, all patients will experience disease progression with this therapy mainly due to the adaptation of cancer cells through primary or secondary molecular mechanisms of resistance. The comprehension and access to tissue and cell-free DNA next-generation sequencing have fueled the development of innovative therapeutic strategies to prevent and overcome resistance to osimertinib in the clinical setting. Herein, we review the biological and clinical implications of molecular mechanisms of osimertinib resistance and the ongoing development of therapeutic strategies to overcome or prevent resistance.
Collapse
Affiliation(s)
- Juan Bautista Blaquier
- Thoracic Oncology Unit, Medical Oncology, Center for Medical Education and Clinical Research (CEMIC), Buenos Aires, Argentina
| | - Sandra Ortiz-Cuaran
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, France
| | - Biagio Ricciuti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Laura Mezquita
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Andrés Felipe Cardona
- Foundation for Clinical and Applied Cancer Research-FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá, Colombia
- Direction of Research and Education, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Cancer-CTIC, Bogotá, Colombia
| | - Gonzalo Recondo
- Thoracic Oncology Unit, Medical Oncology, Center for Medical Education and Clinical Research (CEMIC), Buenos Aires, Argentina
- Medical Oncology Department, Bradford Hill Clinical Research Center, Santiago, Chile
| |
Collapse
|
30
|
Connell W, Garcia K, Goodarzi H, Keiser MJ. Learning chemical sensitivity reveals mechanisms of cellular response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.26.554851. [PMID: 37693536 PMCID: PMC10491110 DOI: 10.1101/2023.08.26.554851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Chemical probes interrogate disease mechanisms at the molecular level by linking genetic changes to observable traits. However, comprehensive chemical screens in diverse biological models are impractical. To address this challenge, we developed ChemProbe, a model that predicts cellular sensitivity to hundreds of molecular probes and drugs by learning to combine transcriptomes and chemical structures. Using ChemProbe, we inferred the chemical sensitivity of cancer cell lines and tumor samples and analyzed how the model makes predictions. We retrospectively evaluated drug response predictions for precision breast cancer treatment and prospectively validated chemical sensitivity predictions in new cellular models, including a genetically modified cell line. Our model interpretation analysis identified transcriptome features reflecting compound targets and protein network modules, identifying genes that drive ferroptosis. ChemProbe is an interpretable in silico screening tool that allows researchers to measure cellular response to diverse compounds, facilitating research into molecular mechanisms of chemical sensitivity.
Collapse
Affiliation(s)
- William Connell
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Kristle Garcia
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Hani Goodarzi
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Michael J. Keiser
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
31
|
Huang M, Wang H, Mackey C, Chung MC, Guan J, Zheng G, Roy A, Xie M, Vulpe C, Tang X. YAP at the Crossroads of Biomechanics and Drug Resistance in Human Cancer. Int J Mol Sci 2023; 24:12491. [PMID: 37569866 PMCID: PMC10419175 DOI: 10.3390/ijms241512491] [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: 07/15/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Biomechanical forces are of fundamental importance in biology, diseases, and medicine. Mechanobiology is an emerging interdisciplinary field that studies how biological mechanisms are regulated by biomechanical forces and how physical principles can be leveraged to innovate new therapeutic strategies. This article reviews state-of-the-art mechanobiology knowledge about the yes-associated protein (YAP), a key mechanosensitive protein, and its roles in the development of drug resistance in human cancer. Specifically, the article discusses three topics: how YAP is mechanically regulated in living cells; the molecular mechanobiology mechanisms by which YAP, along with other functional pathways, influences drug resistance of cancer cells (particularly lung cancer cells); and finally, how the mechanical regulation of YAP can influence drug resistance and vice versa. By integrating these topics, we present a unified framework that has the potential to bring theoretical insights into the design of novel mechanomedicines and advance next-generation cancer therapies to suppress tumor progression and metastasis.
Collapse
Affiliation(s)
- Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Heyang Wang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Cole Mackey
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32603, USA
| | - Michael C. Chung
- Department of Physics, University of Florida, Gainesville, FL 32611, USA
| | - Juan Guan
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
- Department of Physics, University of Florida, Gainesville, FL 32611, USA
| | - Guangrong Zheng
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32603, USA
| | - Arkaprava Roy
- Department of Biostatistics, University of Florida, Gainesville, FL 32603, USA
| | - Mingyi Xie
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32603, USA
| | - Christopher Vulpe
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32603, USA
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
32
|
Li A, Li D, Gu Y, Liu R, Tang X, Zhao Y, Qi F, Wei J, Liu J. Plant-derived nanovesicles: Further exploration of biomedical function and application potential. Acta Pharm Sin B 2023; 13:3300-3320. [PMID: 37655320 PMCID: PMC10465964 DOI: 10.1016/j.apsb.2022.12.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/21/2022] [Accepted: 12/15/2022] [Indexed: 03/09/2023] Open
Abstract
Extracellular vesicles (EVs) are phospholipid bilayer vesicles actively secreted by cells, that contain a variety of functional nucleic acids, proteins, and lipids, and are important mediums of intercellular communication. Based on their natural properties, EVs can not only retain the pharmacological effects of their source cells but also serve as natural delivery carriers. Among them, plant-derived nanovesicles (PNVs) are characterized as natural disease therapeutics with many advantages such as simplicity, safety, eco-friendliness, low cost, and low toxicity due to their abundant resources, large yield, and low risk of immunogenicity in vivo. This review systematically introduces the biogenesis, isolation methods, physical characterization, and components of PNVs, and describes their administration and cellular uptake as therapeutic agents. We highlight the therapeutic potential of PNVs as therapeutic agents and drug delivery carriers, including anti-inflammatory, anticancer, wound healing, regeneration, and antiaging properties as well as their potential use in the treatment of liver disease and COVID-19. Finally, the toxicity and immunogenicity, the current clinical application, and the possible challenges in the future development of PNVs were analyzed. We expect the functions of PNVs to be further explored to promote clinical translation, thereby facilitating the development of a new framework for the treatment of human diseases.
Collapse
Affiliation(s)
- Aixue Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Dan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yongwei Gu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Rongmei Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaomeng Tang
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yunan Zhao
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fu Qi
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jifu Wei
- Department of Pharmacy, Jiangsu Cancer Hospital, Nanjing 210009, China
- Jiangsu Institute of Cancer Research, Nanjing 210009, China
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Jiyong Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pharmacy, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| |
Collapse
|
33
|
Shi YX, Yan JH, Liu W, Deng J. Identifies KCTD5 as a novel cancer biomarker associated with programmed cell death and chemotherapy drug sensitivity. BMC Cancer 2023; 23:408. [PMID: 37149576 PMCID: PMC10163697 DOI: 10.1186/s12885-023-10895-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/27/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND More and more studies have demonstrated that potassium channel tetramerization domain-containing 5 (KCTD5) plays an important role in the development of cancer, but there is a lack of comprehensive research on the biological function of this protein in pan-cancer. This study systematically analyzed the expression landscape of KCTD5 in terms of its correlations with tumor prognosis, the immune microenvironment, programmed cell death, and drug sensitivity. METHODS We investigated a number of databases, including TCGA, GEPIA2, HPA, TISIDB, PrognoScan, GSCA, CellMiner, and TIMER2.0. The study evaluated the expression of KCTD5 in human tumors, as well as its prognostic value and its association with genomic alterations, the immune microenvironment, tumor-associated fibroblasts, functional enrichment analysis, and anticancer drug sensitivity. Real-time quantitative PCR and flow cytometry analysis were performed to determine the biological functions of KCTD5 in lung adenocarcinoma cells. RESULTS The results indicated that KCTD5 is highly expressed in most cancers and that its expression is significantly correlated with tumor prognosis. Moreover, KCTD5 expression was related to the immune microenvironment, infiltration by cancer-associated fibroblasts, and the expression of immune-related genes. Functional enrichment analysis revealed that KCTD5 is associated with apoptosis, necroptosis, and other types of programmed cell death. In vitro experiments showed that knockdown of KCTD5 promoted apoptosis of A549 cells. Correlation analysis confirmed that KCTD5 was positively correlated with the expression of the anti-apoptotic genes Bcl-xL and Mcl-1. Additionally, KCTD5 was significantly associated with sensitivity to multiple antitumor drugs. CONCLUSION Our results suggest that KCTD5 is a potential molecular biomarker that can be used to predict patient prognosis, immunoreactions and drug sensitivity in pan-cancer. KCTD5 plays an important role in regulating programmed cell death, especially apoptosis.
Collapse
Affiliation(s)
- Yuan-Xiang Shi
- Institute of Clinical Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, 410005, P.R. China.
| | - Jian-Hua Yan
- Department of Cardiac Thoracic Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Wen Liu
- Department of Pharmacy, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, 410005, P.R. China
| | - Jun Deng
- Department of Pharmacy, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, 410005, P.R. China
| |
Collapse
|
34
|
Vojtek M, Martins CB, Ramos R, Duarte SG, Ferreira IMPLVO, Batista de Carvalho ALM, Marques MPM, Diniz C. Pd(II) and Pt(II) Trinuclear Chelates with Spermidine: Selective Anticancer Activity towards TNBC-Sensitive and -Resistant to Cisplatin. Pharmaceutics 2023; 15:1205. [PMID: 37111690 PMCID: PMC10145437 DOI: 10.3390/pharmaceutics15041205] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most aggressive forms of breast cancer and constitutes 10-20% of all breast cancer cases. Even though platinum-based drugs such as cisplatin and carboplatin are effective in TNBC patients, their toxicity and development of cancer drug resistance often hamper their clinical use. Hence, novel drug entities with improved tolerability and selectivity profiles, as well as the ability to surpass resistance, are needed. The current study focuses on Pd(II) and Pt(II) trinuclear chelates with spermidine (Pd3Spd2 and Pt3Spd2) for evaluating their antineoplastic activity having been assessed towards (i) cisplatin-resistant TNBC cells (MDA-MB-231/R), (ii) cisplatin-sensitive TNBC cells (MDA-MB-231) and (iii) non-cancerous human breast cells (MCF-12A, to assess the cancer selectivity/selectivity index). Additionally, the complexes' ability to overcome acquired resistance (resistance index) was determined. This study revealed that Pd3Spd2 activity greatly exceeds that displayed by its Pt analog. In addition, Pd3Spd2 evidenced a similar antiproliferative activity in both sensitive and resistant TNBC cells (IC50 values 4.65-8.99 µM and 9.24-13.34 µM, respectively), with a resistance index lower than 2.3. Moreover, this Pd compound showed a promising selectivity index ratio: >6.28 for MDA-MB-231 cells and >4.59 for MDA-MB-231/R cells. Altogether, the data presently gathered reveal Pd3Spd2 as a new, promising metal-based anticancer agent, which should be further explored for the treatment of TNBC and its cisplatin-resistant forms.
Collapse
Affiliation(s)
- Martin Vojtek
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Clara B. Martins
- Molecular Physical-Chemistry R & D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Raquel Ramos
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Sara Gomes Duarte
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Isabel M. P. L. V. O. Ferreira
- LAQV/REQUIMTE, Laboratory of Bromatology and Hydrology, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | | | - M. Paula M. Marques
- Molecular Physical-Chemistry R & D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Carmen Diniz
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
35
|
Miyazaki K, Kishimoto H, Kobayashi H, Suzuki A, Higuchi K, Shirasaka Y, Inoue K. The Glycosylated N-Terminal Domain of MUC1 Is Involved in Chemoresistance by Modulating Drug Permeation Across the Plasma Membrane. Mol Pharmacol 2023; 103:166-175. [PMID: 36804202 DOI: 10.1124/molpharm.122.000597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 11/21/2022] [Indexed: 12/09/2022] Open
Abstract
Mucin 1 (MUC1) is aberrantly expressed in various cancers and implicated in cancer progression and chemoresistance. Although the C-terminal cytoplasmic tail of MUC1 is involved in signal transduction, promoting chemoresistance, the role of the extracellular MUC1 domain [N-terminal glycosylated domain (NG)-MUC1] remains unclear. In this study, we generated stable MCF7 cell lines expressing MUC1 and cytoplasmic tail-deficient MUC1 (MUC1ΔCT) and show that NG-MUC1 is involved in drug resistance by modulating the transmembrane permeation of various compounds without cytoplasmic tail signaling. Heterologous expression of MUC1ΔCT increased cell survival in treating anticancer drugs (such as 5-fluorouracil, cisplatin, doxorubicin, and paclitaxel), in particular by causing an approximately 150-fold increase in the IC50 of paclitaxel, a lipophilic drug, compared with the control [5-fluorouracil (7-fold), cisplatin (3-fold), and doxorubicin (18-fold)]. The uptake studies revealed that accumulations of paclitaxel and Hoechst 33342, a membrane-permeable nuclear staining dye, were reduced to 51% and 45%, respectively, in cells expressing MUC1ΔCT via ABCB1/P-gp-independent mechanisms. Such alterations in chemoresistance and cellular accumulation were not observed in MUC13-expressing cells. Furthermore, we found that MUC1 and MUC1ΔCT increased the cell-adhered water volume by 2.6- and 2.7-fold, respectively, suggesting the presence of a water layer on the cell surface created by NG-MUC1. Taken together, these results suggest that NG-MUC1 acts as a hydrophilic barrier element against anticancer drugs and contributes to chemoresistance by limiting the membrane permeation of lipophilic drugs. Our findings could help better the understanding of the molecular basis of drug resistance in cancer chemotherapy. SIGNIFICANCE STATEMENT: Membrane-bound mucin (MUC1), aberrantly expressed in various cancers, is implicated in cancer progression and chemoresistance. Although the MUC1 cytoplasmic tail is involved in proliferation-promoting signal transduction thereby leading to chemoresistance, the significance of the extracellular domain remains unclear. This study clarifies the role of the glycosylated extracellular domain as a hydrophilic barrier element to limit the cellular uptake of lipophilic anticancer drugs. These findings could help better the understanding of the molecular basis of MUC1 and drug resistance in cancer chemotherapy.
Collapse
Affiliation(s)
- Kaori Miyazaki
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences (K.M., H.Ki, H.Ko, A.S., K.H., and K.I.) and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (Y.S.)
| | - Hisanao Kishimoto
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences (K.M., H.Ki, H.Ko, A.S., K.H., and K.I.) and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (Y.S.)
| | - Hanai Kobayashi
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences (K.M., H.Ki, H.Ko, A.S., K.H., and K.I.) and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (Y.S.)
| | - Ayaka Suzuki
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences (K.M., H.Ki, H.Ko, A.S., K.H., and K.I.) and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (Y.S.)
| | - Kei Higuchi
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences (K.M., H.Ki, H.Ko, A.S., K.H., and K.I.) and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (Y.S.)
| | - Yoshiyuki Shirasaka
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences (K.M., H.Ki, H.Ko, A.S., K.H., and K.I.) and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (Y.S.)
| | - Katsuhisa Inoue
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences (K.M., H.Ki, H.Ko, A.S., K.H., and K.I.) and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (Y.S.)
| |
Collapse
|
36
|
Golan T, Raitses-Gurevich M, Beller T, Carroll J, Brody JR. Strategies for the Management of Patients with Pancreatic Cancer with PARP Inhibitors. Cancer Treat Res 2023; 186:125-142. [PMID: 37978134 DOI: 10.1007/978-3-031-30065-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
A subset of patients with pancreatic adenocarcinomas (PDAC) harbor mutations that are exploitable in the context of DNA-damage response and repair (DDR) inhibitory strategies. Between 8-18% of PDACs harbor specific mutations in the DDR pathway such as BRCA1/2 mutations, and a higher prevalence exists in high-risk populations (e.g., Ashkenazi Jews). Herein, we will review the current trials and data on the treatment of PDAC patients who harbor such mutations and who appear sensitive to platinum and/or poly ADP ribose polymerase inhibitor (PARPi) based therapies due to a concept known as synthetic lethality. Although this current best-in-class precision treatment shows clinical promise, the specter of resistance limits the extent of therapeutic responses. We therefore also evaluate promising pre-clinical and clinical approaches in the pipeline that may either work with existing therapies to break resistance or work separately with combination therapies against this subset of PDACs.
Collapse
Affiliation(s)
- Talia Golan
- Cancer Center, Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Maria Raitses-Gurevich
- Cancer Center, Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Beller
- Cancer Center, Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - James Carroll
- Department of Surgery, Brenden Colson Center for Pancreatic Care, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Jonathan R Brody
- Department of Surgery, Brenden Colson Center for Pancreatic Care, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
37
|
Sola B, Caillot M. L’embryon de poule. Med Sci (Paris) 2022; 38:795-799. [DOI: 10.1051/medsci/2022123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Le développement de drogues anti-cancéreuses à visée thérapeutique nécessite leur évaluation. Ces drogues candidates sont généralement testées in vitro, sur des lignées cellulaires ou sur des cellules isolées à partir de patients, et, in vivo, dans des modèles de xénogreffe chez la souris immunodéprimée. Depuis quelques années, les contraintes réglementaires (règle des 3R : réduire, raffiner, remplacer) imposent de mettre en place des modèles alternatifs qui se substituent aux modèles murins ou, au moins, en limitent l’utilisation. Parmi les modèles alternatifs proposés, la greffe sur membrane chorio-allantoïdienne d’embryon de poule semble performante. Elle permet de suivre et de quantifier la croissance tumorale et d’autres paramètres associés, comme la néo-angiogenèse, l’invasion et la migration tumorales. Elle permet aussi le criblage de drogues. Ce modèle semble également adapté à la médecine personnalisée en cancérologie. Nous présentons dans cette revue la technique et ses avantages.
Collapse
|