1
|
Song H, Xiao S, Wu J, Lu M. Drugging p53: Barriers, Criteria, and Prospects. Cancer Discov 2024; 14:2055-2060. [PMID: 39485253 DOI: 10.1158/2159-8290.cd-24-0837] [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: 07/18/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 11/03/2024]
Abstract
Pharmacologically targeting tumor suppressors necessitates an unprecedented strategy of restoring, rather than conventionally inhibiting, protein function, and p53, the most commonly mutated protein in cancer, has thus remained undruggable. In this study, we address long-standing misconceptions in the field and gaps in the scientific logic for a p53 function-restoration strategy, identify four barriers for drugging mutant p53, and accordingly propose effectiveness evaluation criteria, clinical-translating norms, and prospects for mutant p53 rescue compounds.
Collapse
Affiliation(s)
- Huaxin Song
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujun Xiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaqi Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Lu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Lu J, Chen L, Fatima Z, Huang J, Chen J. Synergistic rescue of temperature-sensitive p53 mutants by hypothermia and arsenic trioxide. Mol Carcinog 2024; 63:2205-2217. [PMID: 39115446 PMCID: PMC11466696 DOI: 10.1002/mc.23804] [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: 06/11/2024] [Revised: 07/15/2024] [Accepted: 07/29/2024] [Indexed: 10/11/2024]
Abstract
The p53 tumor suppressor is inactivated by mutations in about 50% of tumors. Rescuing the transcriptional function of mutant p53 has potential therapeutic benefits. Approximately 15% of p53 mutants are temperature sensitive (TS) and regain maximal activity at 32°C. Proof of concept study showed that induction of 32°C hypothermia in mice restored TS mutant p53 activity and inhibited tumor growth. However, 32°C is the lower limit of therapeutic hypothermia procedures for humans. Higher temperatures are preferable but result in suboptimal TS p53 activation. Recently, arsenic trioxide (ATO) was shown to rescue the conformation of p53 structural mutants by stabilizing the DNA binding domain. We examined the responses of 17 frequently observed p53 TS mutants to functional rescue by temperature shift and ATO. The results showed that ATO only rescued mild p53 TS mutants with high basal activity at 37°C. Mild TS mutants showed a common feature of regaining significant activity at the semi-permissive temperature of 35°C and could be further stimulated by ATO at 35°C. TS p53 rescue by ATO was antagonized by the cellular redox mechanism and was rapidly reversible. Inhibition of glutathione (GSH) biosynthesis enhanced ATO rescue efficiency and sustained p53 activity after ATO washout. The results suggest that mild TS p53 mutants are uniquely responsive to functional rescue by ATO due to small thermostability deficits and inherent potential to regain active conformation. Combining mild hypothermia and ATO may provide an effective and safe procedure for targeting tumors with p53 TS mutations.
Collapse
Affiliation(s)
- Junhao Lu
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Lihong Chen
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Zainab Fatima
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jeffrey Huang
- Department of Anesthesiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jiandong Chen
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| |
Collapse
|
3
|
Puzo CJ, Hager KM, Rinder HM, Weinberg OK, Siddon AJ. Overall survival in TP53-mutated AML and MDS. Ann Hematol 2024:10.1007/s00277-024-06054-7. [PMID: 39443370 DOI: 10.1007/s00277-024-06054-7] [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: 08/08/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
TP53 mutations in patients with AML and MDS frequently portend a poor prognosis, related to both p53 allele status and blast count. In 2022, the ICC and WHO released updated guidelines for classifying p53-mutated AML/MDS. The characteristics of p53 mutations, their associated co-mutations, and their effects on overall survival (OS) are not known in the context of these new guidelines. A retrospective chart review was undertaken for all patients with AML or MDS and at least one TP53 mutation detected on next generation sequencing (NGS) at Yale New Haven Hospital from 2015 to 2023. All patients (N = 210) met criteria for one of the 5 diagnostic classes based on WHO and ICC guidelines. Kaplan-Meier curves with associated log-rank testing and Cox proportional hazards model quantified the effects of clinical and molecular data on survival. Multi-hit pathogenic mutations were related to poorer OS in MDS but not AML using either the WHO (p = .02) or the ICC (p = .01) diagnostic criteria. The most significant predictors of OS in the sample overall were platelet count < 50 K (HR: 2.01, 95% CI [1.47, 2.75], p < .001) and TP53 VAF ≤ 40% (HR: 0.68, 95% CI[0.50, 0.91], p = .01). Blast count ranges, complex karyotype, and p53 mutation type or location, showed no association with OS. In our cohort defined by the 2022 ICC and WHO criteria, VAF and thrombocytopenia, rather than blast count or p53 mutation features, significantly predicted OS. These results speak to each criteria's ability to identify cases of similarly aggressive disease biology and prognosis.
Collapse
Affiliation(s)
| | - Karl M Hager
- Department of Laboratory Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Henry M Rinder
- Department of Laboratory Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Olga K Weinberg
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexa J Siddon
- Department of Laboratory Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
4
|
Hosseini MS. Current insights and future directions of Li-Fraumeni syndrome. Discov Oncol 2024; 15:561. [PMID: 39404911 PMCID: PMC11480288 DOI: 10.1007/s12672-024-01435-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Li-Fraumeni syndrome is a rare yet serious hereditary cancer predisposition syndrome, marked by a significant early-life increased risk of developing cancer. Primarily caused by germline mutations in the TP53 tumor suppressor gene, Li-Fraumeni syndrome is associated with a wide range of malignancies. Clinical management of Li-Fraumeni syndrome could be challenging, especially the lifelong surveillance and follow-up of patients which requires a multidisciplinary approach. Emerging insights into the molecular and clinical basis of Li-Fraumeni syndrome, coupled with advances in genomic technologies and targeted therapies, offer promise in optimizing risk assessment, early detection, and treatment strategies tailored to the unique clinical and molecular profiles of affected individuals. This review discusses Li-Fraumeni syndrome in more depth, reviewing molecular, genomic, epidemiological, clinical, and therapeutic aspects of this disease.
Collapse
Affiliation(s)
- Mohammad-Salar Hosseini
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 51666, EA, Iran.
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Evidence-Based Medicine, Iranian EBM Center: A JBI Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Nzitakera A, Uwamariya D, Kato H, Surwumwe JB, Mbonigaba A, Ndoricyimpaye EL, Uwamungu S, Manirakiza F, Ndayisaba MC, Ntakirutimana G, Seminega B, Dusabejambo V, Rutaganda E, Kamali P, Ngabonziza F, Ishikawa R, Watanabe H, Rugwizangoga B, Baba S, Yamada H, Yoshimura K, Sakai Y, Sugimura H, Shinmura K. TP53 mutation status and consensus molecular subtypes of colorectal cancer in patients from Rwanda. BMC Cancer 2024; 24:1266. [PMID: 39394554 PMCID: PMC11468329 DOI: 10.1186/s12885-024-13009-8] [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: 06/18/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Mutations in the TP53 tumor suppressor gene are well-established drivers of colorectal cancer (CRC) development. However, data on the prevalence of TP53 variants and their association with consensus molecular subtype (CMS) classification in patients with CRC from Rwanda are currently lacking. This study addressed this knowledge gap by investigating TP53 mutation status concerning CMS classification in a CRC cohort from Rwanda. METHODS Formalin-fixed paraffin-embedded (FFPE) tissue blocks were obtained from 51 patients with CRC at the University Teaching Hospital of Kigali, Rwanda. Exons 4 to 11 and their flanking intron-exon boundaries in the TP53 gene were sequenced using Sanger sequencing to identify potential variants. The recently established immunohistochemistry-based classifier was employed to determine the CMS of each tumor. RESULTS Sequencing analysis of cancerous tissue DNA revealed TP53 pathogenic variants in 23 of 51 (45.1%) patients from Rwanda. These variants were predominantly missense types (18/23, 78.3%). The most frequent were c.455dup (p.P153Afs*28), c.524G > A (p.R175H), and c.733G > A (p.G245S), each identified in three tumors. Trinucleotide sequence context analysis of the 23 mutations (20 of which were single-base substitutions) revealed a predominance of the [C > N] pattern among single-base substitutions (SBSs) (18/20; 90.0%), with C[C > T]G being the most frequent mutation (5/18, 27.8%). Furthermore, pyrimidine bases (C and T) were preferentially found at the 5' flanking position of the mutated cytosine (13/18; 72.2%). Analysis of CMS subtypes revealed the following distribution: CMS1 (microsatellite instability-immune) (6/51, 11.8%), CMS2 (canonical) (28/51, 54.9%), CMS3 (metabolic) (9/51, 17.6%), and CMS4 (mesenchymal) (8/51, 15.7%). Interestingly, the majority of TP53 variants were in the CMS2 subgroup (14/23; 60.1%). CONCLUSION Our findings indicate a high frequency of TP53 variants in CRC patients from Rwanda. Importantly, these variants are enriched in the CMS2 subtype. This study, representing the second investigation into molecular alterations in patients with CRC from Rwanda and the first to explore TP53 mutations and CMS classification, provides valuable insights into the molecular landscape of CRC in this understudied population.
Collapse
Affiliation(s)
- Augustin Nzitakera
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
- Department of Biomedical Laboratory Sciences, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Delphine Uwamariya
- Department of Biomedical Laboratory Sciences, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Hisami Kato
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Jean Bosco Surwumwe
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
| | - André Mbonigaba
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Ella Larissa Ndoricyimpaye
- Department of Biomedical Laboratory Sciences, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Université Catholique de Louvain, Médecine Expérimentale, Brussels, 1348, Belgium
| | - Schifra Uwamungu
- Department of Biomedical Laboratory Sciences, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Department of Pharmacology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-40530, Sweden
| | - Felix Manirakiza
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Marie Claire Ndayisaba
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Gervais Ntakirutimana
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Benoit Seminega
- Department of Internal Medicine, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- College of Medicine and Health Sciences, School of Medicine and Pharmacy, University of Rwanda, Kigali, Rwanda
| | - Vincent Dusabejambo
- Department of Internal Medicine, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- College of Medicine and Health Sciences, School of Medicine and Pharmacy, University of Rwanda, Kigali, Rwanda
| | - Eric Rutaganda
- Department of Internal Medicine, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- College of Medicine and Health Sciences, School of Medicine and Pharmacy, University of Rwanda, Kigali, Rwanda
| | - Placide Kamali
- Department of Internal Medicine, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- College of Medicine and Health Sciences, School of Medicine and Pharmacy, University of Rwanda, Kigali, Rwanda
| | - François Ngabonziza
- Department of Internal Medicine, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- College of Medicine and Health Sciences, School of Medicine and Pharmacy, University of Rwanda, Kigali, Rwanda
| | - Rei Ishikawa
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Hirofumi Watanabe
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Belson Rugwizangoga
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Tumor Immunology Laboratory, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, SE- 40530, Sweden
| | - Satoshi Baba
- Department of Diagnostic Pathology, Hamamatsu University School of Medicine, Medicine, 1- 20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Hidetaka Yamada
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Katsuhiro Yoshimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Yasuhiro Sakai
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Haruhiko Sugimura
- Sasaki Institute Sasaki Foundation, 2-2 Kanda Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan.
| | - Kazuya Shinmura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan.
| |
Collapse
|
6
|
Qayoom H, Haq BU, Sofi S, Jan N, Jan A, Mir MA. Targeting mutant p53: a key player in breast cancer pathogenesis and beyond. Cell Commun Signal 2024; 22:484. [PMID: 39390510 PMCID: PMC11466041 DOI: 10.1186/s12964-024-01863-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
The p53 mutation is the most common genetic mutation associated with human neoplasia. TP53 missense mutations, which frequently arise early in breast cancer, are present in over thirty percent of breast tumors. In breast cancer, p53 mutations are linked to a more aggressive course of the disease and worse overall survival rates. TP53 mutations are mostly seen in triple-negative breast cancer, a very diverse kind of the disease. The majority of TP53 mutations originate in the replacement of individual amino acids within the p53 protein's core domain, giving rise to a variety of variations referred to as "mutant p53s." In addition to gaining carcinogenic qualities through gain-of-function pathways, these mutants lose the typical tumor-suppressive features of p53 to variable degrees. The gain-of-function impact of stabilized mutant p53 causes tumor-specific dependency and resistance to therapy. P53 is a prospective target for cancer therapy because of its tumor-suppressive qualities and the numerous alterations that it experiences in tumors. Phenotypic abnormalities in breast cancer, notably poorly differentiated basal-like tumors are frequently linked to high-grade tumors. By comparing data from cell and animal models with clinical outcomes in breast cancer, this study investigates the molecular mechanisms that convert gene alterations into the pathogenic consequences of mutant p53's tumorigenic activity. The study delves into current and novel treatment approaches aimed at targeting p53 mutations, taking into account the similarities and differences in p53 regulatory mechanisms between mutant and wild-type forms, as well.
Collapse
Affiliation(s)
- Hina Qayoom
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India
| | - Burhan Ul Haq
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India
| | - Shazia Sofi
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India
| | - Nusrat Jan
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India
| | - Asma Jan
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India
| | - Manzoor A Mir
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India.
| |
Collapse
|
7
|
Lin YC, Ku CC, Wuputra K, Wu DC, Yokoyama KK. Vulnerability of Antioxidant Drug Therapies on Targeting the Nrf2-Trp53-Jdp2 Axis in Controlling Tumorigenesis. Cells 2024; 13:1648. [PMID: 39404411 PMCID: PMC11475825 DOI: 10.3390/cells13191648] [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: 08/15/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Control of oxidation/antioxidation homeostasis is important for cellular protective functions, and disruption of the antioxidation balance by exogenous and endogenous ligands can lead to profound pathological consequences of cancerous commitment within cells. Although cancers are sensitive to antioxidation drugs, these drugs are sometimes associated with problems including tumor resistance or dose-limiting toxicity in host animals and patients. These problems are often caused by the imbalance between the levels of oxidative stress-induced reactive oxygen species (ROS) and the redox efficacy of antioxidants. Increased ROS levels, because of abnormal function, including metabolic abnormality and signaling aberrations, can promote tumorigenesis and the progression of malignancy, which are generated by genome mutations and activation of proto-oncogene signaling. This hypothesis is supported by various experiments showing that the balance of oxidative stress and redox control is important for cancer therapy. Although many antioxidant drugs exhibit therapeutic potential, there is a heterogeneity of antioxidation functions, including cell growth, cell survival, invasion abilities, and tumor formation, as well as the expression of marker genes including tumor suppressor proteins, cell cycle regulators, nuclear factor erythroid 2-related factor 2, and Jun dimerization protein 2; their effectiveness in cancer remains unproven. Here, we summarize the rationale for the use of antioxidative drugs in preclinical and clinical antioxidant therapy of cancer, and recent advances in this area using cancer cells and their organoids, including the targeting of ROS homeostasis.
Collapse
Affiliation(s)
- Ying-Chu Lin
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Kazunari K. Yokoyama
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
8
|
Lo CS, Alavi P, Bassey-Archibong B, Jahroudi N, Pasdar M. Differential effect of plakoglobin in restoring the tumor suppressor activities of p53-R273H vs. p53-R175H mutants. PLoS One 2024; 19:e0306705. [PMID: 39361615 PMCID: PMC11449273 DOI: 10.1371/journal.pone.0306705] [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: 02/19/2024] [Accepted: 06/21/2024] [Indexed: 10/05/2024] Open
Abstract
The six most common missense mutations in the DNA binding domain of p53 are known as "hot spots" and include two of the most frequently occurring p53 mutations (p53-R175H and p53-R273H). p53 stability and function are regulated by various post-translational modifications such as phosphorylation, acetylation, sumoylation, methylation, and interactions with other proteins including plakoglobin. Previously, using various carcinoma cell lines we showed that plakoglobin interacted with wild-type and several endogenous p53 mutants (e.g., R280K, R273H, S241F, S215R, R175H) and restored their tumor suppressor activities in vitro. Since mutant p53 function is both mutant-specific and cell context-dependent, we sought herein, to determine if plakoglobin tumor suppressive effects on exogenously expressed p53-R273H and p53-R175H mutants are similarly maintained under the same genetic background using the p53-null and plakoglobin-deficient H1299 cell line. Functional assays were performed to assess colony formation, migration, and invasion while immunoblotting and qPCR were used to examine the subcellular distribution and expression of specific proteins and genes that are typically regulated by or regulate p53 function and are altered in mutant p53-expressing cell lines and tumors. We show that though, plakoglobin interacted with both p53-R273H and p53-R175H mutants, it had a differential effect on the transcription and subcellular distribution of their gene targets and their overall oncogenic properties in vitro. Notably, we found that plakoglobin's tumor suppressive effects were significantly stronger in p53-R175H expressing cells compared to p53-R273H cells. Together, our results indicate that exploring plakoglobin interactions with p53-R175H may be useful for the development of cancer therapeutics focused on the restoration of p53 function.
Collapse
Affiliation(s)
- Chu Shiun Lo
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Parnian Alavi
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Blessing Bassey-Archibong
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Biology and Environmental Sciences Concordia University of Edmonton, Edmonton, Alberta, Canada
| | - Nadia Jahroudi
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Manijeh Pasdar
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
9
|
Di Y, Zhang X, Wen X, Qin J, Ye L, Wang Y, Song M, Wang Z, He W. MAPK Signaling-Mediated RFNG Phosphorylation and Nuclear Translocation Restrain Oxaliplatin-Induced Apoptosis and Ferroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402795. [PMID: 39120977 PMCID: PMC11481204 DOI: 10.1002/advs.202402795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/12/2024] [Indexed: 08/11/2024]
Abstract
Chemotherapy resistance remains a major challenge in the treatment of colorectal cancer (CRC). Therefore, it is crucial to develop novel strategies to sensitize cancer cells to chemotherapy. Here, the fringe family is screened to determine their contribution to chemotherapy resistance in CRC. It is found that RFNG depletion significantly sensitizes cancer cells to oxaliplatin treatment. Mechanistically, chemotherapy-activated MAPK signaling induces ERK to phosphorylate RFNG Ser255 residue. Phosphorylated RFNG S255 (pS255) interacts with the nuclear importin proteins KPNA1/importin-α1 and KPNB1/importin-β1, leading to its translocation into the nucleus where it targets p53 and inhibits its phosphorylation by competitively inhibiting the binding of CHK2 to p53. Consequently, the expression of CDKN1A is decreased and that of SLC7A11 is increased, leading to the inhibition of apoptosis and ferroptosis. In contrast, phosphor-deficient RFNG S225A mutant showed increased apoptosis and ferroptosis, and exhibited a notable response to oxaliplatin chemotherapy both in vitro and in vivo. It is further revealed that patients with low RFNG pS255 exhibited significant sensitivity to oxaliplatin in a patient-derived xenograft (PDX) model. These findings highlight the crosstalk between the MAPK and p53 signaling pathways through RFNG, which mediates oxaliplatin resistance in CRC. Additionally, this study provides guidance for oxaliplatin treatment of CRC patients.
Collapse
Affiliation(s)
- Yuqin Di
- Molecular Diagnosis and Gene Testing CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Xiang Zhang
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Xiangqiong Wen
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Jiale Qin
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Lvlan Ye
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Department of BiochemistryZhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Youpeng Wang
- Center of Hepato‐Pancreato‐Biliary SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Mei Song
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510080China
| | - Ziyang Wang
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Weiling He
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Department of Gastrointestinal SurgeryXiang'an Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361000China
| |
Collapse
|
10
|
Guo L, Chen W, Yue J, Gao M, Zhang J, Huang Y, Xiong H, Li X, Wang Y, Yuan Y, Chen L, Fei F, Xu R. Unlocking the potential of LHPP: Inhibiting glioma growth and cell cycle via the MDM2/p53 pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1871:167509. [PMID: 39277057 DOI: 10.1016/j.bbadis.2024.167509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/31/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024]
Abstract
The recurrence of glioma after treatment has remained an intractable problem for many years. Recently, numerous studies have explored the pivotal role of the mouse double minute 2 (MDM2)/p53 pathway in cancer treatment. Lysine phosphate phosphohistidine inorganic pyrophosphate phosphatase (LHPP), a newly discovered tumor suppressor, has been confirmed in numerous studies on tumors, but its role in glioma remains poorly understood. Expression matrices in The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases were analyzed using gene set enrichment analysis (GSEA), revealing significant alterations in the p53 pathway among glioma patients with high LHPP expression. The overexpression of LHPP in glioma cells resulted in a reduction in cell proliferation, migration, and invasive ability, as well as an increase in apoptosis and alterations to the cell cycle. The present study has identified a novel inhibitory mechanism of LHPP against glioma, both in vivo and in vitro. The results demonstrate that LHPP exerts anti-glioma effects via the MDM2/p53 pathway. These findings may offer a new perspective for the treatment of glioma in the clinic.
Collapse
Affiliation(s)
- Lili Guo
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjin Chen
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jiong Yue
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingjun Gao
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jin Zhang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yukai Huang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Huan Xiong
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinda Li
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yangyang Wang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Yuan
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Longyi Chen
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Fan Fei
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
11
|
Dinić J, Jovanović Stojanov S, Dragoj M, Grozdanić M, Podolski-Renić A, Pešić M. Cancer Patient-Derived Cell-Based Models: Applications and Challenges in Functional Precision Medicine. Life (Basel) 2024; 14:1142. [PMID: 39337925 PMCID: PMC11433531 DOI: 10.3390/life14091142] [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: 07/31/2024] [Revised: 08/22/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
The field of oncology has witnessed remarkable progress in personalized cancer therapy. Functional precision medicine has emerged as a promising avenue for achieving superior treatment outcomes by integrating omics profiling and sensitivity testing of patient-derived cancer cells. This review paper provides an in-depth analysis of the evolution of cancer-directed drugs, resistance mechanisms, and the role of functional precision medicine platforms in revolutionizing individualized treatment strategies. Using two-dimensional (2D) and three-dimensional (3D) cell cultures, patient-derived xenograft (PDX) models, and advanced functional assays has significantly improved our understanding of tumor behavior and drug response. This progress will lead to identifying more effective treatments for more patients. Considering the limited eligibility of patients based on a genome-targeted approach for receiving targeted therapy, functional precision medicine provides unprecedented opportunities for customizing medical interventions according to individual patient traits and individual drug responses. This review delineates the current landscape, explores limitations, and presents future perspectives to inspire ongoing advancements in functional precision medicine for personalized cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11108 Belgrade, Serbia; (J.D.); (S.J.S.); (M.D.); (M.G.); (A.P.-R.)
| |
Collapse
|
12
|
Sabile JMG, Swords R, Tyner JW. Evaluating targeted therapies in older patients with TP53-mutated AML. Leuk Lymphoma 2024; 65:1201-1218. [PMID: 38646877 DOI: 10.1080/10428194.2024.2344057] [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: 11/23/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/23/2024]
Abstract
Mutation of thetumor suppressor gene, TP53 (tumor protein 53), occurs in up to 15% of all patients with acute myeloid leukemia (AML) and is enriched within specific clinical subsets, most notably in older adults, and including secondary AML cases arising from preceding myeloproliferative neoplasm (MPN), myelodysplastic syndrome (MDS), patients exposed to prior DNA-damaging, cytotoxic therapies. In all cases, these tumors have remained difficult to effectively treat with conventional therapeutic regimens. Newer approaches fortreatmentofTP53-mutated AML have shifted to interventions that maymodulateTP53 function, target downstream molecular vulnerabilities, target non-p53 dependent molecular pathways, and/or elicit immunogenic responses. This review will describe the basic biology of TP53, the clinical and biological patterns of TP53 within myeloid neoplasms with a focus on elderly AML patients and will summarize newer therapeutic strategies and current clinical trials.
Collapse
Affiliation(s)
- Jean M G Sabile
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Ronan Swords
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey W Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
13
|
Huber A, Allam AH, Dijkstra C, Thiem S, Huynh J, Poh AR, Konecnik J, Jacob SP, Busuttil R, Liao Y, Chisanga D, Shi W, Alorro MG, Forrow S, Tauriello DVF, Batlle E, Boussioutas A, Williams DS, Buchert M, Ernst M, Eissmann MF. Mutant TP53 switches therapeutic vulnerability during gastric cancer progression within interleukin-6 family cytokines. Cell Rep 2024; 43:114616. [PMID: 39128004 PMCID: PMC11372443 DOI: 10.1016/j.celrep.2024.114616] [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: 02/01/2024] [Revised: 06/17/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024] Open
Abstract
Although aberrant activation of the KRAS and PI3K pathway alongside TP53 mutations account for frequent aberrations in human gastric cancers, neither the sequence nor the individual contributions of these mutations have been clarified. Here, we establish an allelic series of mice to afford conditional expression in the glandular epithelium of KrasG12D;Pik3caH1047R or Trp53R172H and/or ablation of Pten or Trp53. We find that KrasG12D;Pik3caH1047R is sufficient to induce adenomas and that lesions progress to carcinoma when also harboring Pten deletions. An additional challenge with either Trp53 loss- or gain-of-function alleles further accelerated tumor progression and triggered metastatic disease. While tumor-intrinsic STAT3 signaling in response to gp130 family cytokines remained as a gatekeeper for all stages of tumor development, metastatic progression required a mutant Trp53-induced interleukin (IL)-11 to IL-6 dependency switch. Consistent with the poorer survival of patients with high IL-6 expression, we identify IL-6/STAT3 signaling as a therapeutic vulnerability for TP53-mutant gastric cancer.
Collapse
Affiliation(s)
- Anne Huber
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Amr H Allam
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Christine Dijkstra
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Stefan Thiem
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Jennifer Huynh
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Joshua Konecnik
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Saumya P Jacob
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Rita Busuttil
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Gastroenterology, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Yang Liao
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Mariah G Alorro
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Stephen Forrow
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Daniele V F Tauriello
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Alex Boussioutas
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Gastroenterology, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - David S Williams
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia; Department of Anatomical Pathology, Austin Health, Heidelberg, VIC 3084, Australia
| | - Michael Buchert
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia.
| | - Moritz F Eissmann
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia.
| |
Collapse
|
14
|
Wang CL, Yang BW, Wang XY, Chen X, Li WD, Zhai HY, Wu Y, Cui MY, Wu JH, Meng QH, Zhang N. Targeting colorectal cancer with Herba Patriniae and Coix seed: Network pharmacology, molecular docking, and in vitro validation. World J Gastrointest Oncol 2024; 16:3539-3558. [PMID: 39171161 PMCID: PMC11334031 DOI: 10.4251/wjgo.v16.i8.3539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Herba Patriniae and Coix seed (HC) constitute a widely utilized drug combination in the clinical management of colorectal cancer (CRC) that is known for its diuretic, anti-inflammatory, and swelling-reducing properties. Although its efficacy has been demonstrated in a clinical setting, the active compounds and their mechanisms of action in CRC treatment remain to be fully elucidated. AIM To identify the active, CRC-targeting components of HC and to elucidate the mechanisms of action involved. METHODS Active HC components were identified and screened using databases. Targets for each component were predicted. CRC-related targets were obtained from human gene databases. Interaction targets between HC and CRC were identified. A "drug-ingredient-target" network was created to identify the core components and targets involved. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were conducted to elucidate the key pathways involved. Molecular docking between core targets and key components was executed. In vitro experiments validated core monomers. RESULTS Nineteen active components of HC were identified, with acacetin as the primary active compound. The predictive analysis identified 454 targets of the active compounds in HC. Intersection mapping with 2685 CRC-related targets yielded 171 intervention targets, including 30 core targets. GO and KEGG analyses indicated that HC may influence the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. Molecular docking showed that acacetin exhibited an optimal interaction with AKT1, identifying PI3K, AKT, and P53 as key genes likely targeted by HC during CRC treatment. Acacetin inhibited HT-29 cell proliferation and migration, as well as promoted apoptosis, in vitro. Western blotting analysis revealed increased p53 and cleaved caspase-3 expression and decreased levels of p-PI3K, p-Akt, and survivin, which likely contributed to CRC apoptosis. CONCLUSION Acacetin, the principal active compound in the HC pair, inhibited the proliferation and migration of HT-29 cells and promoted apoptosis through the PI3K/Akt/p53 signaling pathway.
Collapse
Affiliation(s)
- Cheng-Lei Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang 712046, Shaanxi Province, China
| | - Bing-Wei Yang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xin-Yan Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xue Chen
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Wei-Dong Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Department of Scientific Research Management, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Hao-Yu Zhai
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ying Wu
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang 712046, Shaanxi Province, China
| | - Mu-Yao Cui
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jia-He Wu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Qing-Hui Meng
- School of Clinical Medicine Qinghai University, Xining 810000, Qinghai Province, China
| | - Nan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
15
|
Abbas R, Hartmann O, Asiss DT, Abbas R, Kagan J, Kim HT, Oren M, Diefenbacher M, Orian A, Larisch S. ARTS and small-molecule ARTS mimetics upregulate p53 levels by promoting the degradation of XIAP. Apoptosis 2024; 29:1145-1160. [PMID: 38684550 PMCID: PMC11263447 DOI: 10.1007/s10495-024-01957-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 05/02/2024]
Abstract
Mutations resulting in decreased activity of p53 tumor suppressor protein promote tumorigenesis. P53 protein levels are tightly regulated through the Ubiquitin Proteasome System (UPS). Several E3 ligases were shown to regulate p53 stability, including MDM2. Here we report that the ubiquitin E3 ligase XIAP (X-linked Inhibitors of Apoptosis) is a direct ligase for p53 and describe a novel approach for modulating the levels of p53 by targeting the XIAP pathway. Using in vivo (live-cell) and in vitro (cell-free reconstituted system) ubiquitylation assays, we show that the XIAP-antagonist ARTS regulates the levels of p53 by promoting the degradation of XIAP. XIAP directly binds and ubiquitylates p53. In apoptotic cells, ARTS inhibits the ubiquitylation of p53 by antagonizing XIAP. XIAP knockout MEFs express higher p53 protein levels compared to wild-type MEFs. Computational screen for small molecules with high affinity to the ARTS-binding site within XIAP identified a small-molecule ARTS-mimetic, B3. This compound stimulates apoptosis in a wide range of cancer cells but not normal PBMC (Peripheral Blood Mononuclear Cells). Like ARTS, the B3 compound binds to XIAP and promotes its degradation via the UPS. B3 binding to XIAP stabilizes p53 by disrupting its interaction with XIAP. These results reveal a novel mechanism by which ARTS and p53 regulate each other through an amplification loop to promote apoptosis. Finally, these data suggest that targeting the ARTS binding pocket in XIAP can be used to increase p53 levels as a new strategy for developing anti-cancer therapeutics.
Collapse
Affiliation(s)
- Ruqaia Abbas
- Cell Death and Cancer Research Laboratory, Department of Human Biology and Medical Sciences, University of Haifa, 31905, Haifa, Israel
| | - Oliver Hartmann
- Comprehensive Pneumology Center (CPC)/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Munich, Germany
| | - Dorin Theodora Asiss
- Cell Death and Cancer Research Laboratory, Department of Human Biology and Medical Sciences, University of Haifa, 31905, Haifa, Israel
| | - Rabab Abbas
- Cell Death and Cancer Research Laboratory, Department of Human Biology and Medical Sciences, University of Haifa, 31905, Haifa, Israel
| | - Julia Kagan
- Cell Death and Cancer Research Laboratory, Department of Human Biology and Medical Sciences, University of Haifa, 31905, Haifa, Israel
| | | | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Markus Diefenbacher
- Comprehensive Pneumology Center (CPC)/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Munich, Germany
- Ludwig-Maximilian-Universität München (LMU), Munich, Germany
- German Cancer Consortium (DKTK), LMU, Munich, Germany
| | - Amir Orian
- Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, 3109610, Haifa, Israel
| | - Sarit Larisch
- Cell Death and Cancer Research Laboratory, Department of Human Biology and Medical Sciences, University of Haifa, 31905, Haifa, Israel.
| |
Collapse
|
16
|
Lei Z, Zhu Z, Yao Z, Dai X, Dong Y, Chen B, Wang S, Wang S, Bentum-Ennin L, Jin L, Gu H, Hu W. Reciprocal interactions between lncRNAs and MYC in colorectal cancer: partners in crime. Cell Death Dis 2024; 15:539. [PMID: 39075086 PMCID: PMC11286766 DOI: 10.1038/s41419-024-06918-w] [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: 02/22/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024]
Abstract
Proto-oncogenic MYC is frequently dysregulated in colorectal cancer (CRC). In the past decades, long noncoding RNAs (lncRNAs) have emerged as important regulators in cancers, acting as scaffolds, molecular decoys, post-transcriptional regulators, and others. Interestingly, lncRNAs are able to control MYC expression both at transcriptional and post-transcriptional levels. It is suggested that the reciprocal interaction of MYC and lncRNAs often occurs in CRC. MYC can affect the cell fate by promoting or inhibiting the transcription of some lncRNAs. At the same time, some lncRNAs can also affect MYC expression or transcriptional activity, and in turn decide the cell fate. In this review we summarized the current knowledge about the MYC and lncRNA axis, focusing on its mutual regulation, roles in CRC, and proposed potential therapeutic prospects for CRC treatment.
Collapse
Affiliation(s)
- Zhen Lei
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Zhipu Zhu
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Zhihui Yao
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Xiangyu Dai
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Yi Dong
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Bing Chen
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Songyu Wang
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Siyue Wang
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Lutterodt Bentum-Ennin
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230027, China
| | - Lei Jin
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China.
| | - Hao Gu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230027, China.
| | - Wanglai Hu
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China.
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230027, China.
| |
Collapse
|
17
|
Poonsombudlert K, Mott S, Miller B, Dhakal P, Snow A, Hornberg S, Yodsuwan R, Strouse C, Shaikh H, Magalhaes-Silverman M, Sutamtewagul G. Transplant versus no transplant in myelodysplastic syndrome and acute myeloid leukemia with TP53 mutation; a referral center experience. Eur J Haematol 2024; 113:16-23. [PMID: 38511425 DOI: 10.1111/ejh.14199] [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: 01/03/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/22/2024]
Abstract
A remarkably high rate of post-transplant relapse in patients with TP53-mutated myelodysplastic syndrome/acute myeloid leukemia (MDS/AML) calls to question the utility of allogeneic stem cell transplant (HSCT). We, therefore, performed a retrospective analysis to compare the outcomes between HSCT (N = 38) versus non-HSCT (N = 45) approaches. Patients in the HSCT cohort were younger (median age 63 vs. 72) while patients in the non-HSCT cohort more commonly had complex karyotype with chromosome 17 aberrancy and 5q deletion (p < .01). A total of 69 TP53 variants including 64 pathogenic variants, and 5 variants of undetermined significance were detected. Nine patients (4 in HSCT and 5 in non-HSCT) had multi-hit TP53 variants. After induction: 57.9% versus 56.6% in the HSCT versus non-HSCT cohort achieved morphologic complete remission. Median time to HSCT was 6 months and median follow-up was 15.1 months for HSCT and 5.7 months for non-HSCT. Median disease-free survival (DFS) and overall survival (OS) were 11.7 and 15.9 months for HSCT, and 4.1 and 5.7 months for non-HSCT cohorts, respectively. Non-relapse mortality at 12 months was 22% versus 44% for HSCT versus non-HSCT. In the HSCT cohort, the rate of grade II-IV acute and chronic graft-versus-host disease (GVHD) was 55% and 18%, respectively. None of the patients from the non-HSCT cohort were alive while four patients from the HSCT cohort were alive, in remission, and without GVHD (GRFS) at the time of abstraction. Better treatment strategies for patients with TP53-mutated MDS/AML remain an area of unmet clinical need.
Collapse
MESH Headings
- Humans
- Myelodysplastic Syndromes/therapy
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/mortality
- Myelodysplastic Syndromes/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/diagnosis
- Male
- Middle Aged
- Female
- Mutation
- Hematopoietic Stem Cell Transplantation
- Tumor Suppressor Protein p53/genetics
- Aged
- Retrospective Studies
- Adult
- Transplantation, Homologous
- Treatment Outcome
- Graft vs Host Disease/etiology
- Prognosis
- Aged, 80 and over
Collapse
Affiliation(s)
- Kittika Poonsombudlert
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| | - Sarah Mott
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| | - Benda Miller
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| | - Prajwal Dhakal
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| | - Anthony Snow
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| | - Sarah Hornberg
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| | - Ratdanai Yodsuwan
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| | - Christopher Strouse
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| | - Hira Shaikh
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| | | | - Grerk Sutamtewagul
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| |
Collapse
|
18
|
Qi L, Li G, Li P, Wang H, Fang X, He T, Li J. Twenty years of Gendicine® rAd-p53 cancer gene therapy: The first-in-class human cancer gene therapy in the era of personalized oncology. Genes Dis 2024; 11:101155. [PMID: 38523676 PMCID: PMC10958704 DOI: 10.1016/j.gendis.2023.101155] [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: 05/27/2023] [Revised: 09/09/2023] [Accepted: 10/16/2023] [Indexed: 03/26/2024] Open
Abstract
Genetic mutations in TP53 contribute to human malignancies through various means. To date, there have been a variety of therapeutic strategies targeting p53, including gene therapy to restore normal p53 function, mutant p53 rescue, inhibiting the MDM2-p53 interaction, p53-based vaccines, and a number of other approaches. This review focuses on the functions of TP53 and discusses the aberrant roles of mutant p53 in various types of cancer. Recombinant human p53 adenovirus, trademarked as Gendicine, which is the first anti-tumor gene therapy drug, has made tremendous progress in cancer gene therapy. We herein discuss the biological mechanisms by which Gendicine exerts its effects and describe the clinical responses reported in clinical trials. Notably, the clinical studies suggest that the combination of Gendicine with chemotherapy and/or radiotherapy may produce more pronounced efficacy in slowing tumor growth and progression than gene therapy/chemotherapy alone. Finally, we summarize the methods of administration of recombinant human p53 adenovirus for different cancer types to provide a reference for future clinical trials.
Collapse
Affiliation(s)
- Li Qi
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong 262700, China
| | - Guiqing Li
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong 262700, China
| | - Peipei Li
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong 262700, China
| | - Hongwei Wang
- Decording Therapeutics Corp, Shangha 200000, China
- Yangkun Biogroup Co., Ltd, Nanjing, Jiangsu 210002, China
| | - Xiaolong Fang
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong 262700, China
- Jinming Yu Academician Workstation of Oncology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 262700, China
| | - Tongchuan He
- The University of Chicago, Chicago, IL 60290, USA
| | - Jingjing Li
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong 262700, China
- Jinming Yu Academician Workstation of Oncology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 262700, China
| |
Collapse
|
19
|
Benitez DA, Cumplido-Laso G, Olivera-Gómez M, Del Valle-Del Pino N, Díaz-Pizarro A, Mulero-Navarro S, Román-García A, Carvajal-Gonzalez JM. p53 Genetics and Biology in Lung Carcinomas: Insights, Implications and Clinical Applications. Biomedicines 2024; 12:1453. [PMID: 39062026 PMCID: PMC11274425 DOI: 10.3390/biomedicines12071453] [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: 05/16/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The TP53 gene is renowned as a tumor suppressor, playing a pivotal role in overseeing the cell cycle, apoptosis, and maintaining genomic stability. Dysregulation of p53 often contributes to the initiation and progression of various cancers, including lung cancer (LC) subtypes. The review explores the intricate relationship between p53 and its role in the development and progression of LC. p53, a crucial tumor suppressor protein, exists in various isoforms, and understanding their distinct functions in LC is essential for advancing our knowledge of this deadly disease. This review aims to provide a comprehensive literature overview of p53, its relevance to LC, and potential clinical applications.
Collapse
Affiliation(s)
- Dixan A. Benitez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain; (G.C.-L.); (M.O.-G.); (N.D.V.-D.P.); (A.D.-P.); (S.M.-N.); (A.R.-G.)
| | | | | | | | | | | | | | - Jose Maria Carvajal-Gonzalez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain; (G.C.-L.); (M.O.-G.); (N.D.V.-D.P.); (A.D.-P.); (S.M.-N.); (A.R.-G.)
| |
Collapse
|
20
|
Cui H, Li H, Liu J, Zhao P, Liu Y, Zhong R, Li R, Cheng Y. The predictive value of E2F7 in immunotherapy efficacy for lung adenocarcinoma: An observational study. Medicine (Baltimore) 2024; 103:e38574. [PMID: 38905387 PMCID: PMC11191985 DOI: 10.1097/md.0000000000038574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 05/23/2024] [Indexed: 06/23/2024] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common pathological type of lung cancer. In recent years, immunotherapy has greatly changed the treatment pattern of advanced LUAD. However, only a small proportion of LUAD patients benefitted from immune checkpoint inhibitor therapy. There is an urgent need to develop a biomarker to predict immune therapy response. E2F7 has been shown to be closely related to immune cell infiltration and immune checkpoint expression in tumors. However, it is unclear whether the E2F7 expression is related to the immunotherapy efficacy in LUAD. Therefore, we conducted this study to investigate the clinical characteristics, function, and immunotherapy responsiveness of E2F7 expression, and to explore the potential of E2F7 as an immunotherapy response biomarker in LUAD. We analyzed the clinical characteristics and biological function of E2F7 expression based on data from the Cancer Genome Atlas and Gene Expression Omnibus database. In addition, we used single-cell sequencing data to analyze the immune regulatory effects of E2F7 in LUAD. Furthermore, we analyzed the immunotherapy response prediction ability of E2F7 expression based on the immunotherapy database. Compared to normal lung tissue, E2F7 was specifically overexpressed in LUAD, and its expression was associated with higher malignancy and poor efficacy. E2F7 high expression was an independent risk factor affecting the prognosis of LUAD. E2F7 was enriched in cell division and cell cycle functions. In addition, the expressions of immune checkpoints were correlated with the E2F7 expression. E2F7 was highly expressed in myeloid cells, and E2F7 highly expressed myeloid cells were associated with immune and inflammatory responses. Moreover, the expression level of E2F7 can effectively distinguish different immune therapy responses in LUAD patients. E2F7 was upregulated in LUAD, and high expression of E2F7 was associated with higher malignancy and poor efficacy. E2F7 high expression was an independent risk factor affecting the prognosis of LUAD. Moreover, E2F7 may exert its immunosuppressive effect by affecting the function of myeloid cells. These results indicated the potential role of E2F7 as a biomarker for predicting LUAD immunotherapy responses.
Collapse
Affiliation(s)
- Heran Cui
- Biobank, Jilin Cancer Hospital, Changchun, China
| | - Hui Li
- Biobank, Jilin Cancer Hospital, Changchun, China
- Translational Oncology Research Lab, Jilin Province and Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Jingjing Liu
- Biobank, Jilin Cancer Hospital, Changchun, China
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
| | - Peiyan Zhao
- Translational Oncology Research Lab, Jilin Province and Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Yan Liu
- Translational Oncology Research Lab, Jilin Province and Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Rui Zhong
- Translational Oncology Research Lab, Jilin Province and Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Rixin Li
- Biobank, Jilin Cancer Hospital, Changchun, China
| | - Ying Cheng
- Translational Oncology Research Lab, Jilin Province and Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
| |
Collapse
|
21
|
Chen S, Tse K, Lu Y, Chen S, Tian Y, Tan KT, Li C. Comprehensive genomic profiling and therapeutic implications for Taiwanese patients with treatment-naïve breast cancer. Cancer Med 2024; 13:e7384. [PMID: 38895905 PMCID: PMC11187859 DOI: 10.1002/cam4.7384] [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: 11/06/2023] [Revised: 03/29/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Breast cancer is a heterogeneous disease categorized based on molecular characteristics, including hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2) expression levels. The emergence of profiling technology has revealed multiple driver genomic alterations within each breast cancer subtype, serving as biomarkers to predict treatment outcomes. This study aimed to explore the genomic landscape of breast cancer in the Taiwanese population through comprehensive genomic profiling (CGP) and identify diagnostic and predictive biomarkers. METHODS Targeted next-generation sequencing-based CGP was performed on 116 archived Taiwanese breast cancer specimens, assessing genomic alterations (GAs), including single nucleotide variants, copy number variants, fusion genes, tumor mutation burden (TMB), and microsatellite instability (MSI) status. Predictive variants for FDA-approved therapies were evaluated within each subtype. RESULTS In the cohort, frequent mutations included PIK3CA (39.7%), TP53 (36.2%), KMT2C (9.5%), GATA3 (8.6%), and SF3B1 (6.9%). All subtypes had low TMB, with no MSI-H tumors. Among HR + HER2- patients, 42% (27/65) harbored activating PIK3CA mutations, implying potential sensitivity to PI3K inhibitors and resistance to endocrine therapies. HR + HER2- patients exhibited intrinsic hormonal resistance via FGFR1 gene gain/amplification (15%), exclusive of PI3K/AKT pathway alterations. Aberrations in the PI3K/AKT/mTOR and FGFR pathways were implicated in chemoresistance, with a 52.9% involvement in triple-negative breast cancer. In HER2+ tumors, 50% harbored GAs potentially conferring resistance to anti-HER2 therapies, including PIK3CA mutations (32%), MAP3K1 (2.9%), NF1 (2.9%), and copy number gain/amplification of FGFR1 (18%), FGFR3 (2.9%), EGFR (2.9%), and AKT2 (2.9%). CONCLUSION This study presents CGP findings for treatment-naïve Taiwanese breast cancer, emphasizing its value in routine breast cancer management, disease classification, and treatment selection.
Collapse
Affiliation(s)
- Shang‐Hung Chen
- National Institute of Cancer Research, National Health Research InstitutesTainanTaiwan
- Department of OncologyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
| | | | | | | | - Yu‐Feng Tian
- Division of Colorectal Surgery, Department of SurgeryChi Mei Medical CenterTainanTaiwan
- Department of Health and NutritionChia‐Nan University of Pharmacy and ScienceTainanTaiwan
| | - Kien Thiam Tan
- ACT Genomics, Co. Ltd.TaipeiTaiwan
- Anbogen Therapeutics, Inc.TaipeiTaiwan
| | - Chien‐Feng Li
- National Institute of Cancer Research, National Health Research InstitutesTainanTaiwan
- Department of Medical ResearchChi Mei Medical CenterTainanTaiwan
- Institute of Precision MedicineNational Sun Yat‐Sen UniversityKaohsiungTaiwan
- Department of Clinical Pathology and Laboratory MedicineChi Mei Medical CenterTainanTaiwan
- Trans‐omic Laboratory for Precision MedicineChi Mei Medical CenterTainanTaiwan
| |
Collapse
|
22
|
Ohki R, Itahana K, Iwakuma T. "The 10th International MDM2 Workshop": Opening up new avenues for MDM2 and p53 research, the First International MDM2 Workshop in Asia. Genes Cells 2024; 29:451-455. [PMID: 38553254 PMCID: PMC11164626 DOI: 10.1111/gtc.13114] [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: 01/15/2024] [Revised: 02/25/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024]
Abstract
The 10th International MDM2 Workshop was held at the National Cancer Center Research Institute (NCCRI) in Tokyo, Japan, from October 15 to 18, 2023. It attracted 166 participants from 12 countries. The meeting featured 52 talks and 41 poster presentations. In the first special session, six invited speakers gave educational and outstanding talks on breakthroughs in MDM2 research. Three keynote speakers presented emerging p53-independent functions of MDM2/MDM4, functional association of MDM2/p53 with cancer immunity, and drug discovery targeting the MDM2/MDM4-p53 pathway. Additionally, 19 invited speakers introduced their new findings. Twenty-one presenters, many of whom were young investigators, postdocs, and students, were selected from submitted abstracts and reported their exciting and unpublished results. For poster presenters, outstanding poster awards were given to the best presenters. There were many inspiring questions and discussions throughout the meeting. Social events like a welcome party, a workshop dinner, and an optional tour enabled further scientific interactions among the participants. The meeting successfully provided an exciting platform for scientific exchange. The experience gained from organizing this meeting will be handed over to the next organizers of the 11th International MDM2 Workshop.
Collapse
Affiliation(s)
- Rieko Ohki
- Laboratory of Fundamental Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Koji Itahana
- Cancer & Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Tomoo Iwakuma
- Children’s Mercy Research Institute, Kansas City, MO, USA
| |
Collapse
|
23
|
Guo X, Zhang Y, Li Q, Shi F, HuangFu Y, Li J, Lao X. The influence of a modified p53 C-terminal peptide by using a tumor-targeting sequence on cellular apoptosis and tumor treatment. Apoptosis 2024; 29:865-881. [PMID: 38145442 DOI: 10.1007/s10495-023-01926-1] [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] [Accepted: 12/01/2023] [Indexed: 12/26/2023]
Abstract
The restoration of the function of p53 in tumors is a therapeutic strategy for the highly frequent mutation of the TP53 tumor suppressor gene. P460 is a wild-type peptide derived from the p53 C-terminus and has been proven to be capable of restoring the tumor suppressor function of p53. The poor accumulation of drugs in tumors is a serious hindrance to tumor treatment. For enhancing the activity of P460, the tumor-targeting sequence Arg-Gly-Asp-Arg (RGDR, C-end rule peptide) was introduced into the C-terminus of P460 to generate the new peptide P462. P462 presented better activity than P460 in inhibiting the proliferation of cancer cells and increasing the number of tumor cells undergoing apoptosis. Cell adhesion analysis and tumor imaging results revealed that P462 showed more specific and extensive binding with tumor cells and greater accumulation in tumors than the wild-type peptide. Importantly, treatment with P462 was more efficacious than that with P460 in vivo and was associated with considerably improved tumor-homing activity. This study highlights the importance of the roles of the tumor-homing sequence RGDR in the enhancement in cell attachment and tumor accumulation. The results of this work indicate that P462 could be a novel drug candidate for tumor treatment.
Collapse
Affiliation(s)
- Xiaoye Guo
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China
| | - Yiming Zhang
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China
| | - Qian Li
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China
| | - Fangxin Shi
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China
| | - Yifan HuangFu
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China
| | - Jing Li
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China.
| | - Xingzhen Lao
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China.
| |
Collapse
|
24
|
Wang K, Jiang M, Liu H, Meng C, Li M, Lu H. Discovery of novel co-degradation CK1α and CDK7/9 PROTACs with p53 activation for treating acute myeloid leukemia. Bioorg Chem 2024; 147:107319. [PMID: 38593529 DOI: 10.1016/j.bioorg.2024.107319] [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: 01/14/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024]
Abstract
Reactivating p53 activity to restore its anticancer function is an attractive cancer treatment strategy. In this study, we designed and synthesized a series of novel PROTACs to reactivate p53 via the co-degradation of CK1α and CDK7/9 proteins. Bioactivity studies showed that the selected PROTAC 13i exhibited potency antiproliferative activity in MV4-11 (IC50 = 0.096 ± 0.012 μM) and MOLM-13 (IC50 = 0.072 ± 0.014 μM) cells, and induced apoptosis of MV4-11 cells. Western-blot analysis showed that PROTAC 13i triple CK1α and CDK7/9 protein degradation resulted in the significantly increased expression of p53. At the same time, the transcriptional repression due to the degradation significantly reduced downstream gene expression of MYC, MDM2, BCL-2 and MCL-1, and reduced the inflammatory cytokine levels of TNF-α, IL-1β and IL-6 in PMBCs. These results indicate the beneficial impact of simultaneous CK1α and CDK7/9 degradation for acute myeloid leukemia therapy.
Collapse
MESH Headings
- Humans
- Tumor Suppressor Protein p53/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Casein Kinase Ialpha/metabolism
- Casein Kinase Ialpha/antagonists & inhibitors
- Cell Proliferation/drug effects
- Drug Screening Assays, Antitumor
- Cyclin-Dependent Kinase 9/antagonists & inhibitors
- Cyclin-Dependent Kinase 9/metabolism
- Structure-Activity Relationship
- Molecular Structure
- Cyclin-Dependent Kinases/antagonists & inhibitors
- Cyclin-Dependent Kinases/metabolism
- Dose-Response Relationship, Drug
- Apoptosis/drug effects
- Drug Discovery
- Cell Line, Tumor
- Proteolysis/drug effects
- Tumor Cells, Cultured
- Proteolysis Targeting Chimera
- Cyclin-Dependent Kinase-Activating Kinase
Collapse
Affiliation(s)
- Kai Wang
- College of Pharmacy, Jilin University, Changchun 130021, China
| | - Meixu Jiang
- College of Pharmacy, Jilin University, Changchun 130021, China
| | - Huimin Liu
- College of Pharmacy, Jilin University, Changchun 130021, China
| | - Chen Meng
- College of Pharmacy, Jilin University, Changchun 130021, China
| | - Mengyuan Li
- College of Pharmacy, Jilin University, Changchun 130021, China
| | - Haibin Lu
- College of Pharmacy, Jilin University, Changchun 130021, China.
| |
Collapse
|
25
|
Wu HH, Leng S, Sergi C, Leng R. How MicroRNAs Command the Battle against Cancer. Int J Mol Sci 2024; 25:5865. [PMID: 38892054 PMCID: PMC11172831 DOI: 10.3390/ijms25115865] [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: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
MicroRNAs (miRNAs) are small RNA molecules that regulate more than 30% of genes in humans. Recent studies have revealed that miRNAs play a crucial role in tumorigenesis. Large sets of miRNAs in human tumors are under-expressed compared to normal tissues. Furthermore, experiments have shown that interference with miRNA processing enhances tumorigenesis. Multiple studies have documented the causal role of miRNAs in cancer, and miRNA-based anticancer therapies are currently being developed. This review primarily focuses on two key points: (1) miRNAs and their role in human cancer and (2) the regulation of tumor suppressors by miRNAs. The review discusses (a) the regulation of the tumor suppressor p53 by miRNA, (b) the critical role of the miR-144/451 cluster in regulating the Itch-p63-Ago2 pathway, and (c) the regulation of PTEN by miRNAs. Future research and the perspectives of miRNA in cancer are also discussed. Understanding these pathways will open avenues for therapeutic interventions targeting miRNA regulation.
Collapse
Affiliation(s)
- Hong Helena Wu
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada;
| | - Sarah Leng
- Department of Laboratory Medicine and Pathology (5B4. 09), University of Alberta, Edmonton, AB T6G 2B7, Canada (C.S.)
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology (5B4. 09), University of Alberta, Edmonton, AB T6G 2B7, Canada (C.S.)
- Division of Anatomical Pathology, Children’s Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
| | - Roger Leng
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada;
| |
Collapse
|
26
|
Hsu CL, Chang YS, Li HP. Molecular Diagnosis of Nasopharyngeal Carcinoma: Past and Future. Biomed J 2024:100748. [PMID: 38796105 DOI: 10.1016/j.bj.2024.100748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/01/2024] [Accepted: 05/12/2024] [Indexed: 05/28/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor originated from the nasopharynx epithelial cells and has been linked with Epstein-Barr virus (EBV) infection, dietary habits, environmental and genetic factors. It is a common malignancy in Southeast Asia, especially with gender preference among men. Due to its non-specific symptoms, NPC is often diagnosed at a late stage. Thus, the molecular diagnosis of NPC plays a crucial role in early detection, treatment selection, disease monitoring, and prognosis prediction. This review aims to provide a summary of the current state and the latest emerging molecular diagnostic techniques for NPC, including EBV-related biomarkers, gene mutations, liquid biopsy, and DNA methylation. Challenges and potential future directions of NPC molecular diagnosis will be discussed.
Collapse
Affiliation(s)
- Cheng-Lung Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan; School of Medicine, Chang Gung University, Taoyuan 33305, Taiwan.
| | - Yu-Sun Chang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33305, Taiwan; Molecular Medicine Research Center, Chang Gung University, Taoyuan 33305, Taiwan; Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan.
| | - Hsin-Pai Li
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33305, Taiwan; Molecular Medicine Research Center, Chang Gung University, Taoyuan 33305, Taiwan; Department of Microbiology and Immunology, Chang Gung University, Taoyuan 33305, Taiwan.
| |
Collapse
|
27
|
Puccio S, Azzarello G, Maffeis V, Laurino L, Mairani E, Conte F, Tessari N, Cazzador D, Zanoletti E, Politi D, Emanuelli E, Spinato G, Ausoni S. Tumor Budding, p53, and DNA Mismatch Repair Markers in Sinonasal Intestinal-Type Adenocarcinoma: A Retrospective Study Supports the Adverse Prognostic Impact of Tumor Budding. Cancers (Basel) 2024; 16:1895. [PMID: 38791973 PMCID: PMC11120584 DOI: 10.3390/cancers16101895] [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: 03/28/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Sinonasal intestinal-type adenocarcinoma (ITAC) is a very rare, closely occupational-related tumor with strong histological similarities to colorectal cancer (CRC). In the latter, tumor budding (TB) is widely recognized as a negative prognostic parameter. The aim of this study was to evaluate the prognostic role of TB in ITAC and to correlate it with other established or emerging biomarkers of the disease, such as p53 and deficient DNA mismatch repair (MMR) system status/microsatellite instability (MSI). We retrospectively analyzed 32 consecutive specimens of patients with ITAC diagnosis treated in two institutions in Northern Italy. We reviewed surgical specimens for TB evaluation (low-intermediate/high); p53 expression and MMR proteins were evaluated via immunohistochemistry. Results were retrospectively stratified using clinical data and patients' outcomes. According to bud counts, patients were stratified into two groups: intermediate/high budding (>4 TB) and low budding (≤4 TB). Patients with high TB (>4) have an increased risk of recurrence and death compared to those with low TB, with a median survival of 13 and 54 months, respectively. On multivariate analysis, considering TB, therapy, and stage as covariates, TB emerged as an independent prognostic factor net of the stage of disease or type of therapy received. No impact of p53 status as a biomarker of prognosis was observed and no alterations regarding MMR proteins were identified. The results of the present work provide further significant evidence on the prognostic role of TB in ITAC and underline the need for larger multicenter studies to implement the use of TB in clinical practice.
Collapse
Affiliation(s)
- Sebastiano Puccio
- Section of Otorhinolaryngology—Head and Neck Surgery, Department of Neuroscience, “Azienda Ospedale Università di Padova” University of Padova, via Giustiniani, 2, 35122 Padova, Italy; (S.P.); (E.M.); (N.T.); (D.C.); (E.Z.)
| | - Giuseppe Azzarello
- Department of Oncology, Local Health Unit 3 Serenissima, Via don Giacobbe Sartor 4, 30035 Venice, Italy;
| | - Valeria Maffeis
- Anatomia Patologica, Azienda Ospedaliera Universitaria Integrata Verona (AOVR), Ospedale Borgo Trento, Piazzale Aristide Stefani, 1, 37126 Verona, Italy;
- Department of Pathology, Local Health Unit 2 Marca Trevigiana, Piazzale dell’Ospedale 1, 31100 Treviso, Italy;
| | - Licia Laurino
- Department of Pathology, Local Health Unit 3 Serenissima, Via Paccagnella 11, 30174 Venice, Italy;
| | - Edoardo Mairani
- Section of Otorhinolaryngology—Head and Neck Surgery, Department of Neuroscience, “Azienda Ospedale Università di Padova” University of Padova, via Giustiniani, 2, 35122 Padova, Italy; (S.P.); (E.M.); (N.T.); (D.C.); (E.Z.)
| | - Federica Conte
- Department of Psychology, University of Milano-Bicocca, 20126 Milan, Italy;
| | - Nicola Tessari
- Section of Otorhinolaryngology—Head and Neck Surgery, Department of Neuroscience, “Azienda Ospedale Università di Padova” University of Padova, via Giustiniani, 2, 35122 Padova, Italy; (S.P.); (E.M.); (N.T.); (D.C.); (E.Z.)
| | - Diego Cazzador
- Section of Otorhinolaryngology—Head and Neck Surgery, Department of Neuroscience, “Azienda Ospedale Università di Padova” University of Padova, via Giustiniani, 2, 35122 Padova, Italy; (S.P.); (E.M.); (N.T.); (D.C.); (E.Z.)
| | - Elisabetta Zanoletti
- Section of Otorhinolaryngology—Head and Neck Surgery, Department of Neuroscience, “Azienda Ospedale Università di Padova” University of Padova, via Giustiniani, 2, 35122 Padova, Italy; (S.P.); (E.M.); (N.T.); (D.C.); (E.Z.)
| | - Doriano Politi
- Department of Otorhinolaryngology, Local Health Unit 3 Serenissima, Via Paccagnella, 11, 30174 Venice, Italy;
| | - Enzo Emanuelli
- Department of Pathology, Local Health Unit 2 Marca Trevigiana, Piazzale dell’Ospedale 1, 31100 Treviso, Italy;
| | - Giacomo Spinato
- Section of Otorhinolaryngology—Head and Neck Surgery, Department of Neuroscience, “Azienda Ospedale Università di Padova” University of Padova, via Giustiniani, 2, 35122 Padova, Italy; (S.P.); (E.M.); (N.T.); (D.C.); (E.Z.)
| | - Simonetta Ausoni
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58b, 35121 Padova, Italy
| |
Collapse
|
28
|
Olaoba OT, Adelusi TI, Yang M, Maidens T, Kimchi ET, Staveley-O’Carroll KF, Li G. Driver Mutations in Pancreatic Cancer and Opportunities for Targeted Therapy. Cancers (Basel) 2024; 16:1808. [PMID: 38791887 PMCID: PMC11119842 DOI: 10.3390/cancers16101808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Pancreatic cancer is the sixth leading cause of cancer-related mortality globally. As the most common form of pancreatic cancer, pancreatic ductal adenocarcinoma (PDAC) represents up to 95% of all pancreatic cancer cases, accounting for more than 300,000 deaths annually. Due to the lack of early diagnoses and the high refractory response to the currently available treatments, PDAC has a very poor prognosis, with a 5-year overall survival rate of less than 10%. Targeted therapy and immunotherapy are highly effective and have been used for the treatment of many types of cancer; however, they offer limited benefits in pancreatic cancer patients due to tumor-intrinsic and extrinsic factors that culminate in drug resistance. The identification of key factors responsible for PDAC growth and resistance to different treatments is highly valuable in developing new effective therapeutic strategies. In this review, we discuss some molecules which promote PDAC initiation and progression, and their potential as targets for PDAC treatment. We also evaluate the challenges associated with patient outcomes in clinical trials and implications for future research.
Collapse
Affiliation(s)
- Olamide T. Olaoba
- Department of Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA; (O.T.O.); (T.I.A.); (M.Y.); (E.T.K.)
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Temitope I. Adelusi
- Department of Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA; (O.T.O.); (T.I.A.); (M.Y.); (E.T.K.)
| | - Ming Yang
- Department of Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA; (O.T.O.); (T.I.A.); (M.Y.); (E.T.K.)
| | - Tessa Maidens
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA;
| | - Eric T. Kimchi
- Department of Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA; (O.T.O.); (T.I.A.); (M.Y.); (E.T.K.)
| | - Kevin F. Staveley-O’Carroll
- Department of Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA; (O.T.O.); (T.I.A.); (M.Y.); (E.T.K.)
| | - Guangfu Li
- Department of Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA; (O.T.O.); (T.I.A.); (M.Y.); (E.T.K.)
| |
Collapse
|
29
|
Abdulbaki R, Pullarkat ST. A Brief Overview of the Molecular Landscape of Myelodysplastic Neoplasms. Curr Oncol 2024; 31:2353-2363. [PMID: 38785456 PMCID: PMC11119831 DOI: 10.3390/curroncol31050175] [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: 03/13/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Myelodysplastic neoplasm (MDS) is a heterogeneous group of clonal hematological disorders that originate from the hematopoietic and progenitor cells and present with cytopenias and morphologic dysplasia with a propensity to progress to bone marrow failure or acute myeloid leukemia (AML). Genetic evolution plays a critical role in the pathogenesis, progression, and clinical outcomes of MDS. This process involves the acquisition of genetic mutations in stem cells that confer a selective growth advantage, leading to clonal expansion and the eventual development of MDS. With the advent of next-generation sequencing (NGS) assays, an increasing number of molecular aberrations have been discovered in recent years. The knowledge of molecular events in MDS has led to an improved understanding of the disease process, including the evolution of the disease and prognosis, and has paved the way for targeted therapy. The 2022 World Health Organization (WHO) Classification and the International Consensus Classification (ICC) have incorporated the molecular signature into the classification system for MDS. In addition, specific germline mutations are associated with MDS development, especially in pediatrics and young adults. This article reviews the genetic abnormalities of MDS in adults with a brief review of germline predisposition syndromes.
Collapse
Affiliation(s)
- Rami Abdulbaki
- Department of Pathology, Laboratory Medicine, UCLA, David Geffen School of Medicine, Los Angeles, CA 90095, USA;
| | | |
Collapse
|
30
|
Zhang X, Ma H, Gao Y, Liang Y, Du Y, Hao S, Ni T. The Tumor Microenvironment: Signal Transduction. Biomolecules 2024; 14:438. [PMID: 38672455 PMCID: PMC11048169 DOI: 10.3390/biom14040438] [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: 03/06/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
In the challenging tumor microenvironment (TME), tumors coexist with diverse stromal cell types. During tumor progression and metastasis, a reciprocal interaction occurs between cancer cells and their environment. These interactions involve ongoing and evolving paracrine and proximal signaling. Intrinsic signal transduction in tumors drives processes such as malignant transformation, epithelial-mesenchymal transition, immune evasion, and tumor cell metastasis. In addition, cancer cells embedded in the tumor microenvironment undergo metabolic reprogramming. Their metabolites, serving as signaling molecules, engage in metabolic communication with diverse matrix components. These metabolites act as direct regulators of carcinogenic pathways, thereby activating signaling cascades that contribute to cancer progression. Hence, gaining insights into the intrinsic signal transduction of tumors and the signaling communication between tumor cells and various matrix components within the tumor microenvironment may reveal novel therapeutic targets. In this review, we initially examine the development of the tumor microenvironment. Subsequently, we delineate the oncogenic signaling pathways within tumor cells and elucidate the reciprocal communication between these pathways and the tumor microenvironment. Finally, we give an overview of the effect of signal transduction within the tumor microenvironment on tumor metabolism and tumor immunity.
Collapse
Affiliation(s)
- Xianhong Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.L.); (Y.D.)
| | - Haijun Ma
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, China;
| | - Yue Gao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.L.); (Y.D.)
| | - Yabing Liang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.L.); (Y.D.)
| | - Yitian Du
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.L.); (Y.D.)
| | - Shuailin Hao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.L.); (Y.D.)
| | - Ting Ni
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.L.); (Y.D.)
| |
Collapse
|
31
|
Pereira MP, Herrity E, Kim DDH. TP53-mutated acute myeloid leukemia and myelodysplastic syndrome: biology, treatment challenges, and upcoming approaches. Ann Hematol 2024; 103:1049-1067. [PMID: 37770618 DOI: 10.1007/s00277-023-05462-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/17/2023] [Indexed: 09/30/2023]
Abstract
Improved understanding of TP53 biology and the clinicopathological features of TP53-mutated myeloid neoplasms has led to the recognition of TP53-mutated acute myeloid leukemia/myelodysplastic syndrome (TP53m AML/MDS) as a unique entity, characterized by dismal outcomes following conventional therapies. Several clinical trials have investigated combinations of emerging therapies for these patients with the poorest molecular prognosis among myeloid neoplasms. Although some emerging therapies have shown improvement in overall response rates, this has not translated into better overall survival, hence the notion that p53 remains an elusive target. New therapeutic strategies, including novel targeted therapies, immune checkpoint inhibitors, and monoclonal antibodies, represent a shift away from cytotoxic and hypomethylating-based therapies, towards approaches combining non-immune and novel immune therapeutic strategies. The triple combination of azacitidine and venetoclax with either magrolimab or eprenetapopt have demonstrated safety in early trials, with phase III trials currently underway, and promising interim clinical results. This review compiles background on TP53 biology, available and emerging therapies along with their mechanisms of action for the TP53m disease entity, current treatment challenges, and recently published data and status of ongoing clinical trials for TP53m AML/MDS.
Collapse
Affiliation(s)
- Mariana Pinto Pereira
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada
| | - Elizabeth Herrity
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada
| | - Dennis D H Kim
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada.
- Leukemia Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
- Department of Hematology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
32
|
Wang H, Wang Z, Wang Z, Li X, Li Y, Yan N, Wu L, Liang Y, Wu J, Song H, Qu Q, Huang J, Chang C, Shen K, Chen X, Lu M. Decitabine induces IRF7-mediated immune responses in p53-mutated triple-negative breast cancer: a clinical and translational study. Front Med 2024; 18:357-374. [PMID: 38157193 DOI: 10.1007/s11684-023-1016-8] [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] [Received: 12/20/2022] [Accepted: 06/24/2023] [Indexed: 01/03/2024]
Abstract
p53 is mutated in half of cancer cases. However, no p53-targeting drugs have been approved. Here, we reposition decitabine for triple-negative breast cancer (TNBC), a subtype with frequent p53 mutations and extremely poor prognosis. In a retrospective study on tissue microarrays with 132 TNBC cases, DNMT1 overexpression was associated with p53 mutations (P = 0.037) and poor overall survival (OS) (P = 0.010). In a prospective DEciTabinE and Carboplatin in TNBC (DETECT) trial (NCT03295552), decitabine with carboplatin produced an objective response rate (ORR) of 42% in 12 patients with stage IV TNBC. Among the 9 trialed patients with available TP53 sequencing results, the 6 patients with p53 mutations had higher ORR (3/6 vs. 0/3) and better OS (16.0 vs. 4.0 months) than the patients with wild-type p53. In a mechanistic study, isogenic TNBC cell lines harboring DETECT-derived p53 mutations exhibited higher DNMT1 expression and decitabine sensitivity than the cell line with wild-type p53. In the DETECT trial, decitabine induced strong immune responses featuring the striking upregulation of the innate immune player IRF7 in the p53-mutated TNBC cell line (upregulation by 16-fold) and the most responsive patient with TNBC. Our integrative studies reveal the potential of repurposing decitabine for the treatment of p53-mutated TNBC and suggest IRF7 as a potential biomarker for decitabine-based treatments.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhengyuan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zheng Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyang Li
- Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuntong Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ni Yan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lili Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Liang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiale Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Huaxin Song
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qing Qu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiahui Huang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chunkang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200025, China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Min Lu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
33
|
Neri P, Boise LH. TP53 function over forms in multiple myeloma. Blood 2024; 143:1202-1204. [PMID: 38546638 DOI: 10.1182/blood.2023023487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
|
34
|
Lajmi N, Alves-Vasconcelos S, Tsiachristas A, Haworth A, Woods K, Crichton C, Noble T, Salih H, Várnai KA, Branford-White H, Orrell L, Osman A, Bradley KM, Bonney L, McGowan DR, Davies J, Prime MS, Hassan AB. Challenges and solutions to system-wide use of precision oncology as the standard of care paradigm. CAMBRIDGE PRISMS. PRECISION MEDICINE 2024; 2:e4. [PMID: 38699518 PMCID: PMC11062796 DOI: 10.1017/pcm.2024.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/02/2024] [Accepted: 03/12/2024] [Indexed: 05/05/2024]
Abstract
The personalised oncology paradigm remains challenging to deliver despite technological advances in genomics-based identification of actionable variants combined with the increasing focus of drug development on these specific targets. To ensure we continue to build concerted momentum to improve outcomes across all cancer types, financial, technological and operational barriers need to be addressed. For example, complete integration and certification of the 'molecular tumour board' into 'standard of care' ensures a unified clinical decision pathway that both counteracts fragmentation and is the cornerstone of evidence-based delivery inside and outside of a research setting. Generally, integrated delivery has been restricted to specific (common) cancer types either within major cancer centres or small regional networks. Here, we focus on solutions in real-world integration of genomics, pathology, surgery, oncological treatments, data from clinical source systems and analysis of whole-body imaging as digital data that can facilitate cost-effectiveness analysis, clinical trial recruitment, and outcome assessment. This urgent imperative for cancer also extends across the early diagnosis and adjuvant treatment interventions, individualised cancer vaccines, immune cell therapies, personalised synthetic lethal therapeutics and cancer screening and prevention. Oncology care systems worldwide require proactive step-changes in solutions that include inter-operative digital working that can solve patient centred challenges to ensure inclusive, quality, sustainable, fair and cost-effective adoption and efficient delivery. Here we highlight workforce, technical, clinical, regulatory and economic challenges that prevent the implementation of precision oncology at scale, and offer a systematic roadmap of integrated solutions for standard of care based on minimal essential digital tools. These include unified decision support tools, quality control, data flows within an ethical and legal data framework, training and certification, monitoring and feedback. Bridging the technical, operational, regulatory and economic gaps demands the joint actions from public and industry stakeholders across national and global boundaries.
Collapse
Affiliation(s)
- Nesrine Lajmi
- Diagnostics Division, Roche Information Solutions, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Sofia Alves-Vasconcelos
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Apostolos Tsiachristas
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, Oxford, UK
| | - Andrew Haworth
- Diagnostics Division, Roche Information Solutions, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Kerrie Woods
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Theresa Noble
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Hizni Salih
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Kinga A. Várnai
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Liam Orrell
- Diagnostics Division, Roche Information Solutions, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Andrew Osman
- Roche Healthcare Consulting, Roche Diagnostics Limited, West Sussex, UK
| | - Kevin M. Bradley
- Wales Research and Diagnostic PET Imaging Centre, University Hospital of Wales, Cardiff, UK
| | - Lara Bonney
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Jim Davies
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Oxford, UK
| | - Matthew S. Prime
- Diagnostics Division, Roche Information Solutions, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Andrew Bassim Hassan
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
35
|
Alruwaili MM, Zonneville J, Naranjo MN, Serio H, Melendy T, Straubinger RM, Gillard B, Foster BA, Rajan P, Attwood K, Chatley S, Iyer R, Fountzilas C, Bakin AV. A synergistic two-drug therapy specifically targets a DNA repair dysregulation that occurs in p53-deficient colorectal and pancreatic cancers. Cell Rep Med 2024; 5:101434. [PMID: 38387463 PMCID: PMC10982975 DOI: 10.1016/j.xcrm.2024.101434] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 12/06/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024]
Abstract
The tumor-suppressor p53 is commonly inactivated in colorectal cancer and pancreatic ductal adenocarcinoma, but existing treatment options for p53-mutant (p53Mut) cancer are largely ineffective. Here, we report a therapeutic strategy for p53Mut tumors based on abnormalities in the DNA repair response. Investigation of DNA repair upon challenge with thymidine analogs reveals a dysregulation in DNA repair response in p53Mut cells that leads to accumulation of DNA breaks. Thymidine analogs do not interrupt DNA synthesis but induce DNA repair that involves a p53-dependent checkpoint. Inhibitors of poly(ADP-ribose) polymerase (PARPis) markedly enhance DNA double-strand breaks and cell death induced by thymidine analogs in p53Mut cells, whereas p53 wild-type cells respond with p53-dependent inhibition of the cell cycle. Combinations of trifluorothymidine and PARPi agents demonstrate superior anti-neoplastic activity in p53Mut cancer models. These findings support a two-drug combination strategy to improve outcomes for patients with p53Mut cancer.
Collapse
Affiliation(s)
- Mohammed M Alruwaili
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Medical Laboratory Technology, College of Applied Medical Science, Northern Border University, Arar City, Saudi Arabia
| | - Justin Zonneville
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Maricris N Naranjo
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hannah Serio
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Thomas Melendy
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, NY 14214, USA
| | - Robert M Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA; Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Bryan Gillard
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Barbara A Foster
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Priyanka Rajan
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Sarah Chatley
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Renuka Iyer
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Christos Fountzilas
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Andrei V Bakin
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
36
|
Zhan W, Hu H, Hao B, Zhu H, Yan T, Zhang J, Wang S, Liu S, Zhang T. Development of machine learning-based malignant pericardial effusion-related model in breast cancer: Implications for clinical significance, tumor immune and drug-therapy. Heliyon 2024; 10:e27507. [PMID: 38463870 PMCID: PMC10923851 DOI: 10.1016/j.heliyon.2024.e27507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/30/2024] [Accepted: 02/29/2024] [Indexed: 03/12/2024] Open
Abstract
Background Malignant pericardial effusion (MPE) is a common complication of advanced breast cancer (BRCA) and plays an important role in BRCA. This study is aims to construct a prognostic model based on MPE-related genes for predicting the prognosis of breast cancer. Methods The BRCA samples are analyzed based on the expression of MPE-related genes by using an unsupervised cluster analysis method. This study processes the data by least absolute shrinkage and selection operator and multivariate Cox analysis, and uses machine learning algorithms to construct BRCA prognostic model and develop web tool. Results BRCA patients are classified into three clusters and a BRCA prognostic model is constructed containing 9 MPE-related genes. There are significant differences in signature pathways, immune infiltration, immunotherapy response and drug sensitivity testing between the high and low-risk groups. Of note, a web-based tool (http://wys.helyly.top/cox.html) is developed to predict overall survival as well as drug-therapy response of BRCA patients quickly and conveniently, which can provide a basis for clinicians to formulate individualized treatment plans. Conclusion The MPE-related prognostic model developed in this study can be used as an effective tool for predicting the prognosis of BRCA and provides new insights for the diagnosis and treatment of BRCA patients.
Collapse
Affiliation(s)
- Wendi Zhan
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan, 421001, China
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Haihong Hu
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan, 421001, China
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Bo Hao
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hongxia Zhu
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan, 421001, China
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Ting Yan
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jingdi Zhang
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan, 421001, China
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Siyu Wang
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Saiyang Liu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Taolan Zhang
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Phase I Clinical Trial Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
37
|
Timofeev O, Giron P, Lawo S, Pichler M, Noeparast M. ERK pathway agonism for cancer therapy: evidence, insights, and a target discovery framework. NPJ Precis Oncol 2024; 8:70. [PMID: 38485987 PMCID: PMC10940698 DOI: 10.1038/s41698-024-00554-5] [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: 11/03/2023] [Accepted: 02/16/2024] [Indexed: 03/18/2024] Open
Abstract
At least 40% of human cancers are associated with aberrant ERK pathway activity (ERKp). Inhibitors targeting various effectors within the ERKp have been developed and explored for over two decades. Conversely, a substantial body of evidence suggests that both normal human cells and, notably to a greater extent, cancer cells exhibit susceptibility to hyperactivation of ERKp. However, this vulnerability of cancer cells remains relatively unexplored. In this review, we reexamine the evidence on the selective lethality of highly elevated ERKp activity in human cancer cells of varying backgrounds. We synthesize the insights proposed for harnessing this vulnerability of ERK-associated cancers for therapeutical approaches and contextualize these insights within established pharmacological cancer-targeting models. Moreover, we compile the intriguing preclinical findings of ERK pathway agonism in diverse cancer models. Lastly, we present a conceptual framework for target discovery regarding ERKp agonism, emphasizing the utilization of mutual exclusivity among oncogenes to develop novel targeted therapies for precision oncology.
Collapse
Affiliation(s)
- Oleg Timofeev
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35043, Marburg, Germany
| | - Philippe Giron
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research group Genetics, Reproduction and Development, Centre for Medical Genetics, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Steffen Lawo
- CRISPR Screening Core Facility, Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Martin Pichler
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany
| | - Maxim Noeparast
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany.
| |
Collapse
|
38
|
Xiao S, Shi F, Song H, Cui J, Zheng D, Zhang H, Tan K, Wu J, Chen X, Wu J, Tang Y, Dai Y, Lu M. Characterization of the generic mutant p53-rescue compounds in a broad range of assays. Cancer Cell 2024; 42:325-327. [PMID: 38402608 DOI: 10.1016/j.ccell.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/30/2023] [Accepted: 01/17/2024] [Indexed: 02/27/2024]
Abstract
Dozens of compounds that rescue tumor-associated mutant p53 have been reported. Xiao et al. perform 10 assays to evaluate effectiveness of the mutant p53-rescue compounds side-by-side but do not detect reliable rescue in any assay for the evaluated compounds, except for ATO and its analog PAT.
Collapse
Affiliation(s)
- Shujun Xiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Fangfang Shi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huaxin Song
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyi Cui
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Derun Zheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hesong Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Tan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaqi Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueqin Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiale Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yigang Tang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Min Lu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
39
|
Snaith O, Poveda-Rogers C, Laczko D, Yang G, Morrissette JJD. Cytogenetics and genomics of acute myeloid leukemia. Best Pract Res Clin Haematol 2024; 37:101533. [PMID: 38490763 DOI: 10.1016/j.beha.2023.101533] [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: 02/28/2023] [Revised: 09/14/2023] [Accepted: 12/03/2023] [Indexed: 03/17/2024]
Abstract
The diversity of genetic and genomic abnormalities observed in acute myeloid leukemia (AML) reflects the complexity of these hematologic neoplasms. The detection of cytogenetic and molecular alterations is fundamental to diagnosis, risk stratification and treatment of AML. Chromosome rearrangements are well established in the diagnostic classification of AML, as are some gene mutations, in several international classification systems. Additionally, the detection of new mutational profiles at relapse and identification of mutations in the pre- and post-transplant settings are illuminating in understanding disease evolution and are relevant to the risk assessment of AML patients. In this review, we discuss recurrent cytogenetic abnormalities, as well as the detection of recurrent mutations, within the context of a normal karyotype, and in the setting of chromosome abnormalities. Two new classification schemes from the WHO and ICC are described, comparing these classifications in terms of diagnostic criteria and entity definition in AML. Finally, we discuss ways in which genomic sequencing can condense the detection of gene mutations and chromosome abnormalities into a single assay.
Collapse
Affiliation(s)
- Oraine Snaith
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Corey Poveda-Rogers
- Division of Precision and Computational Diagnostics, Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Dorottya Laczko
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Guang Yang
- Division of Precision and Computational Diagnostics, Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer J D Morrissette
- Division of Precision and Computational Diagnostics, Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Peuget S, Zhou X, Selivanova G. Translating p53-based therapies for cancer into the clinic. Nat Rev Cancer 2024; 24:192-215. [PMID: 38287107 DOI: 10.1038/s41568-023-00658-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/31/2024]
Abstract
Inactivation of the most important tumour suppressor gene TP53 occurs in most, if not all, human cancers. Loss of functional wild-type p53 is achieved via two main mechanisms: mutation of the gene leading to an absence of tumour suppressor activity and, in some cases, gain-of-oncogenic function; or inhibition of the wild-type p53 protein mediated by overexpression of its negative regulators MDM2 and MDMX. Because of its high potency as a tumour suppressor and the dependence of at least some established tumours on its inactivation, p53 appears to be a highly attractive target for the development of new anticancer drugs. However, p53 is a transcription factor and therefore has long been considered undruggable. Nevertheless, several innovative strategies have been pursued for targeting dysfunctional p53 for cancer treatment. In mutant p53-expressing tumours, the predominant strategy is to restore tumour suppressor function with compounds acting either in a generic manner or otherwise selective for one or a few specific p53 mutations. In addition, approaches to deplete mutant p53 or to target vulnerabilities created by mutant p53 expression are currently under development. In wild-type p53 tumours, the major approach is to protect p53 from the actions of MDM2 and MDMX by targeting these negative regulators with inhibitors. Although the results of at least some clinical trials of MDM2 inhibitors and mutant p53-restoring compounds are promising, none of the agents has yet been approved by the FDA. Alternative strategies, based on a better understanding of p53 biology, the mechanisms of action of compounds and treatment regimens as well as the development of new technologies are gaining interest, such as proteolysis-targeting chimeras for MDM2 degradation. Other approaches are taking advantage of the progress made in immune-based therapies for cancer. In this Review, we present these ongoing clinical trials and emerging approaches to re-evaluate the current state of knowledge of p53-based therapies for cancer.
Collapse
Affiliation(s)
- Sylvain Peuget
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Xiaolei Zhou
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Institute of Materials Science and Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Galina Selivanova
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
41
|
Song B, Yang P, Zhang S. Cell fate regulation governed by p53: Friends or reversible foes in cancer therapy. Cancer Commun (Lond) 2024; 44:297-360. [PMID: 38311377 PMCID: PMC10958678 DOI: 10.1002/cac2.12520] [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: 07/26/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Cancer is a leading cause of death worldwide. Targeted therapies aimed at key oncogenic driver mutations in combination with chemotherapy and radiotherapy as well as immunotherapy have benefited cancer patients considerably. Tumor protein p53 (TP53), a crucial tumor suppressor gene encoding p53, regulates numerous downstream genes and cellular phenotypes in response to various stressors. The affected genes are involved in diverse processes, including cell cycle arrest, DNA repair, cellular senescence, metabolic homeostasis, apoptosis, and autophagy. However, accumulating recent studies have continued to reveal novel and unexpected functions of p53 in governing the fate of tumors, for example, functions in ferroptosis, immunity, the tumor microenvironment and microbiome metabolism. Among the possibilities, the evolutionary plasticity of p53 is the most controversial, partially due to the dizzying array of biological functions that have been attributed to different regulatory mechanisms of p53 signaling. Nearly 40 years after its discovery, this key tumor suppressor remains somewhat enigmatic. The intricate and diverse functions of p53 in regulating cell fate during cancer treatment are only the tip of the iceberg with respect to its equally complicated structural biology, which has been painstakingly revealed. Additionally, TP53 mutation is one of the most significant genetic alterations in cancer, contributing to rapid cancer cell growth and tumor progression. Here, we summarized recent advances that implicate altered p53 in modulating the response to various cancer therapies, including chemotherapy, radiotherapy, and immunotherapy. Furthermore, we also discussed potential strategies for targeting p53 as a therapeutic option for cancer.
Collapse
Affiliation(s)
- Bin Song
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Ping Yang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Shuyu Zhang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
- The Second Affiliated Hospital of Chengdu Medical CollegeChina National Nuclear Corporation 416 HospitalChengduSichuanP. R. China
- Laboratory of Radiation MedicineNHC Key Laboratory of Nuclear Technology Medical TransformationWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
42
|
Naeimzadeh Y, Tajbakhsh A, Fallahi J. Understanding the prion-like behavior of mutant p53 proteins in triple-negative breast cancer pathogenesis: The current therapeutic strategies and future directions. Heliyon 2024; 10:e26260. [PMID: 38390040 PMCID: PMC10881377 DOI: 10.1016/j.heliyon.2024.e26260] [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: 10/23/2023] [Revised: 01/20/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
Breast cancer (BC) is viewed as a significant public health issue and is the primary cause of cancer-related deaths among women worldwide. Triple-negative breast cancer (TNBC) is a particularly aggressive subtype that predominantly affects young premenopausal women. The tumor suppressor p53 playsa vital role in the cellular response to DNA damage, and its loss or mutations are commonly present in many cancers, including BC. Recent evidence suggests that mutant p53 proteins can aggregate and form prion-like structures, which may contribute to the pathogenesis of different types of malignancies, such as BC. This review provides an overview of BC molecular subtypes, the epidemiology of TNBC, and the role of p53 in BC development. We also discuss the potential implications of prion-like aggregation in BC and highlight future research directions. Moreover, a comprehensive analysis of the current therapeutic approaches targeting p53 aggregates in BC treatment is presented. Strategies including small molecules, chaperone inhibitors, immunotherapy, CRISPR-Cas9, and siRNA are discussed, along with their potential benefits and drawbacks. The use of these approaches to inhibit p53 aggregation and degradation represents a promising target for cancer therapy. Future investigations into the efficacy of these approaches against various p53 mutations or binding to non-p53 proteins should be conducted to develop more effective and personalized therapies for BC treatment.
Collapse
Affiliation(s)
- Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| |
Collapse
|
43
|
Cai HQ, Zhang LY, Fu LM, Xu B, Jiao Y. Mutational landscape of TP53 and CDH1 in gastric cancer. World J Gastrointest Surg 2024; 16:276-283. [PMID: 38463349 PMCID: PMC10921187 DOI: 10.4240/wjgs.v16.i2.276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/26/2023] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
In this editorial we comment on an article published in a recent issue of the World J Gastrointest Surg. A common gene mutation in gastric cancer (GC) is the TP53 mutation. As a tumor suppressor gene, TP53 is implicated in more than half of all tumor occurrences. TP53 gene mutations in GC tissue may be related with clinical pathological aspects. The TP53 mutation arose late in the progression of GC and aided in the final switch to malignancy. CDH1 encodes E-cadherin, which is involved in cell-to-cell adhesion, epithelial structure maintenance, cell polarity, differentiation, and intracellular signaling pathway modulation. CDH1 mutations and functional loss can result in diffuse GC, and CDH1 mutations can serve as independent prognostic indicators for poor prognosis. GC patients can benefit from genetic counseling and testing for CDH1 mutations. Demethylation therapy may assist to postpone the onset and progression of GC. The investigation of TP53 and CDH1 gene mutations in GC allows for the investigation of the relationship between these two gene mutations, as well as providing some basis for evaluating the prognosis of GC patients.
Collapse
Affiliation(s)
- Hong-Qiao Cai
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Li-Yue Zhang
- Department of Critical Care Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Li-Ming Fu
- Department of Traditional Chinese Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Bin Xu
- Department of Traditional Chinese Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
44
|
Wang D, Yan K, Yu H, Li H, Zhou W, Hong Y, Guo S, Wang Y, Xu C, Pan C, Tang Y, Liu N, Wu W, Zhang L, Xi Q. Fimepinostat Impairs NF-κB and PI3K/AKT Signaling and Enhances Gemcitabine Efficacy in H3.3K27M-Diffuse Intrinsic Pontine Glioma. Cancer Res 2024; 84:598-615. [PMID: 38095539 DOI: 10.1158/0008-5472.can-23-0394] [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: 02/07/2023] [Revised: 06/26/2023] [Accepted: 12/07/2023] [Indexed: 02/16/2024]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is the most aggressive pediatric brain tumor, and the oncohistone H3.3K27M mutation is associated with significantly worse clinical outcomes. Despite extensive research efforts, effective approaches for treating DIPG are lacking. Through drug screening, we identified the combination of gemcitabine and fimepinostat as a potent therapeutic intervention for H3.3K27M DIPG. H3.3K27M facilitated gemcitabine-induced apoptosis in DIPG, and gemcitabine stabilized and activated p53, including increasing chromatin accessibility for p53 at apoptosis-related loci. Gemcitabine simultaneously induced a prosurvival program in DIPG through activation of RELB-mediated NF-κB signaling. Specifically, gemcitabine induced the transcription of long terminal repeat elements, activated cGAS-STING signaling, and stimulated noncanonical NF-κB signaling. A drug screen in gemcitabine-treated DIPG cells revealed that fimepinostat, a dual inhibitor of HDAC and PI3K, effectively suppressed the gemcitabine-induced NF-κB signaling in addition to blocking PI3K/AKT activation. Combination therapy comprising gemcitabine and fimepinostat elicited synergistic antitumor effects in vitro and in orthotopic H3.3K27M DIPG xenograft models. Collectively, p53 activation using gemcitabine and suppression of RELB-mediated NF-κB activation and PI3K/AKT signaling using fimepinostat is a potential therapeutic strategy for treating H3.3K27M DIPG. SIGNIFICANCE Gemcitabine activates p53 and induces apoptosis to elicit antitumor effects in H3.3K27M DIPG, which can be enhanced by blocking NF-κB and PI3K/AKT signaling with fimepinostat, providing a synergistic combination therapy for DIPG.
Collapse
Affiliation(s)
- Dan Wang
- MOE Key Laboratory of Protein Sciences, State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Kun Yan
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Hongxing Yu
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Haocheng Li
- MOE Key Laboratory of Protein Sciences, State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Wei Zhou
- MOE Key Laboratory of Protein Sciences, State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yaqiang Hong
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Shuning Guo
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Cheng Xu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Changcun Pan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yujie Tang
- Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nian Liu
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Wei Wu
- MOE Key Laboratory of Protein Sciences, State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing, China
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Beijing Key Laboratory of Brain Tumor, Beijing, China
| | - Qiaoran Xi
- MOE Key Laboratory of Protein Sciences, State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing, China
- Joint Graduate Program of Peking-Tsinghua-NIBS, Tsinghua University, Beijing, China
| |
Collapse
|
45
|
Liu Z, Yang Y, Sun X, Ma R, Zhang W, Wang W, Yang G, Wang H, Zhang J, Wang Y, Tian J. Discovery of Novel Antitumor Small-Molecule Agent with Dual Action of CDK2/p-RB and MDM2/p53. Molecules 2024; 29:725. [PMID: 38338471 PMCID: PMC10856454 DOI: 10.3390/molecules29030725] [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: 01/03/2024] [Revised: 01/26/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Cell cycle-dependent kinase 2 (CDK2) is located downstream of CDK4/6 in the cell cycle and regulates cell entry into S-phase by binding to Cyclin E and hyper-phosphorylating Rb. Proto-oncogene murine double minute 2 (MDM2) is a key negative regulator of p53, which is highly expressed in tumors and plays an important role in tumorigenesis and progression. In this study, we identified a dual inhibitor of CDK2 and MDM2, III-13, which had good selectivity for inhibiting CDK2 activity and significantly reduced MDM2 expression. In vitro results showed that III-13 inhibited proliferation of a wide range of tumor cells, regardless of whether Cyclin E1 (CCNE1) was overexpressed or not. The results of in vivo experiments showed that III-13 significantly inhibited proliferation of tumor cells and did not affect body weight of mice. The results of the druggability evaluation showed that III-13 was characterized by low bioavailability and poor membrane permeability when orally administered, suggesting the necessity of further structural modifications. Therefore, this study provided a lead compound for antitumor drugs, especially those against CCNE1-amplified tumor proliferation.
Collapse
Affiliation(s)
- Zhaofeng Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (Z.L.); (Y.Y.); (X.S.); (R.M.); (W.W.); (G.Y.); (H.W.); (J.Z.)
| | - Yifei Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (Z.L.); (Y.Y.); (X.S.); (R.M.); (W.W.); (G.Y.); (H.W.); (J.Z.)
| | - Xiaohui Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (Z.L.); (Y.Y.); (X.S.); (R.M.); (W.W.); (G.Y.); (H.W.); (J.Z.)
| | - Runchen Ma
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (Z.L.); (Y.Y.); (X.S.); (R.M.); (W.W.); (G.Y.); (H.W.); (J.Z.)
| | - Wenjing Zhang
- R & D Center, Luye Pharma Group Ltd., Yantai 264003, China;
| | - Wenyan Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (Z.L.); (Y.Y.); (X.S.); (R.M.); (W.W.); (G.Y.); (H.W.); (J.Z.)
| | - Gangqiang Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (Z.L.); (Y.Y.); (X.S.); (R.M.); (W.W.); (G.Y.); (H.W.); (J.Z.)
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (Z.L.); (Y.Y.); (X.S.); (R.M.); (W.W.); (G.Y.); (H.W.); (J.Z.)
| | - Jianzhao Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (Z.L.); (Y.Y.); (X.S.); (R.M.); (W.W.); (G.Y.); (H.W.); (J.Z.)
| | - Yunjie Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (Z.L.); (Y.Y.); (X.S.); (R.M.); (W.W.); (G.Y.); (H.W.); (J.Z.)
| | - Jingwei Tian
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (Z.L.); (Y.Y.); (X.S.); (R.M.); (W.W.); (G.Y.); (H.W.); (J.Z.)
| |
Collapse
|
46
|
Stockhammer P, Grant M, Wurtz A, Foggetti G, Expósito F, Gu J, Zhao H, Choi J, Chung S, Li F, Walther Z, Dietz J, Duffield E, Gettinger S, Politi K, Goldberg SB. Co-Occurring Alterations in Multiple Tumor Suppressor Genes Are Associated With Worse Outcomes in Patients With EGFR-Mutant Lung Cancer. J Thorac Oncol 2024; 19:240-251. [PMID: 37806385 PMCID: PMC11364167 DOI: 10.1016/j.jtho.2023.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/10/2023] [Accepted: 10/01/2023] [Indexed: 10/10/2023]
Abstract
INTRODUCTION Patients with metastatic EGFR-mutant NSCLC inevitably have disease progression while on tyrosine kinase inhibitor (TKI) therapy. Co-occurring tumor suppressor gene (TSG) alterations have been associated with poor outcomes, however, detailed analyses of their impact on patient outcomes are limited. METHODS Patients with EGFR-mutant NSCLC treated with EGFR TKIs who had tumor genomic profiling were included. Alterations in TP53 and five additional TSGs (RB1, NF1, ARID1A, BRCA1, and PTEN) were used to stratify the cohort into the following three subgroups: patients with tumors harboring a TP53 mutation plus a mutation in at least one additional TSG (TP53mut/TSGmut), those having a TP53 mutation without additional TSG mutations (TP53mut/TSGwt), and those with TP53wt. Patient characteristics and clinical outcomes were assessed in two independent cohorts. RESULTS A total of 101 patients from the Yale Cancer Center and 182 patients from the American Association for Cancer Research Project GENIE database were included. In the Yale cohort, TP53 mutations were identified in 65 cases (64%), of which 23 were TP53mut/TSGmut and 42 were TP53mut/TSGwt. Although the presence of a TP53 mutation was associated with worse outcomes, the additional TSG alteration in TP53mut tumors identified a subset of patients associated with particularly aggressive disease and inferior clinical outcome in both the Yale and the GENIE cohorts. Specifically, in the Yale cohort for patients receiving first-line TKIs, those with TP53mut/TSGmut tumors had shorter progression-free survival (PFS) and overall survival (OS) than TP53mut/TSGwt (PFS: hazard ratio [HR] = 2.03, confidence interval [CI]: 1.12-3.69, p < 0.01, OS: HR = 1.58, CI: 0.82-3.04, p = 0.12) or TP53wt cases (PFS: HR 2.4, CI: 1.28-4.47, p < 0.001, OS: HR = 2.54, CI: 1.21-5.34, p < 0.005). Inferior outcomes in patients with TP53mut/TSGmut tumors were also found in those receiving osimertinib as second-line therapy. Similar findings were seen in patients in the GENIE cohort. CONCLUSIONS Patients with TP53mut/TSGmut tumors represent a patient subgroup characterized by an aggressive disease phenotype and inferior outcomes on EGFR TKIs. This information is important for understanding the biological underpinnings of differential outcomes with TKI treatment and has implications for identifying patients who may benefit from additional therapeutic interventions beyond osimertinib monotherapy.
Collapse
Affiliation(s)
- Paul Stockhammer
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Michael Grant
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Anna Wurtz
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Giorgia Foggetti
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut; Vita-Salute San Raffaele University, Milano, Italy; Medical Oncology Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milano, Italy
| | - Francisco Expósito
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Jianlei Gu
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
| | - Sangyun Chung
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Fangyong Li
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Zenta Walther
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Julia Dietz
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Emily Duffield
- Yale New Haven Hospital, Smilow Cancer Hospital, New Haven, Connecticut
| | - Scott Gettinger
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Katerina Politi
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut; Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Sarah B Goldberg
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut.
| |
Collapse
|
47
|
Mei Y, Qin X, Yang Z, Song S, Liu X, Wu C, Qian J, Huang X, Zhang Y, He W. Engineered a dual-targeting HA-TPP/A nanoparticle for combination therapy against KRAS-TP53 co-mutation in gastrointestinal cancers. Bioact Mater 2024; 32:277-291. [PMID: 37876556 PMCID: PMC10590736 DOI: 10.1016/j.bioactmat.2023.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/19/2023] [Accepted: 10/03/2023] [Indexed: 10/26/2023] Open
Abstract
KRAS-TP53 co-mutation is strongly associated with poor prognosis and high malignancy in gastrointestinal cancers. Therefore, a novel approach to oncotherapy may lie in combination therapy targeting both KRAS and TP53. Herein, we present a novel self-assembled nanoparticle (HA-TPP/A) that are functionalized nano-carrier hyaluronic acid (HA)-TPP conjugate (HA-TPP) to degrade mutant p53 proteins (mutp53) and co-deliver AMG510 for treating KRAS-TP53 co-alteration of gastrointestinal cancers by inhibiting the mutant KRAS and mutp53 signaling pathways. The HA-TPP/A nanoparticles led to ubiquitination-dependent proteasomal degradation of mutp53 by targeting damage to mitochondria. Furthermore, these nanoparticles abrogated the gain-of-function (GOF) phenotypes of mutp53 and increased sensitivity to AMG510-induced cell killing, thereby reducing cell proliferation and migration in gastrointestinal cancer with KRAS-TP53 co-mutation. The co-loaded HA-TPP/A nanoparticles demonstrated remarkable therapeutic efficacy in a tumor-bearing mouse model, particularly in KRAS-TP53 double mutant expressing cancer cells, compared with single drug and combined free drug groups. Notably, HA-TPP/A is the first reported nanoparticle with an ability to co-target KRAS-TP53, providing a promising approach for therapy in highly malignant gastrointestinal tumors and potentially expanding clinical indications for AMG510 targeted therapies in gastrointestinal tumors.
Collapse
Affiliation(s)
- Yong Mei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaohua Qin
- School of Biomedical Science and Engineering, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Zhenyu Yang
- School of Biomedical Science and Engineering, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Shiyao Song
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaoting Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Chong Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jieying Qian
- School of Biomedical Science and Engineering, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Xiaowan Huang
- School of Biomedical Science and Engineering, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Yunjiao Zhang
- School of Biomedical Science and Engineering, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction and Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Weiling He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China
| |
Collapse
|
48
|
Tuval A, Strandgren C, Heldin A, Palomar-Siles M, Wiman KG. Pharmacological reactivation of p53 in the era of precision anticancer medicine. Nat Rev Clin Oncol 2024; 21:106-120. [PMID: 38102383 DOI: 10.1038/s41571-023-00842-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2023] [Indexed: 12/17/2023]
Abstract
p53, which is encoded by the most frequently mutated gene in cancer, TP53, is an attractive target for novel cancer therapies. Despite major challenges associated with this approach, several compounds that either augment the activity of wild-type p53 or restore all, or some, of the wild-type functions to p53 mutants are currently being explored. In wild-type TP53 cancer cells, p53 function is often abrogated by overexpression of the negative regulator MDM2, and agents that disrupt p53-MDM2 binding can trigger a robust p53 response, albeit potentially with induction of p53 activity in non-malignant cells. In TP53-mutant cancer cells, compounds that promote the refolding of missense mutant p53 or the translational readthrough of nonsense mutant TP53 might elicit potent cell death. Some of these compounds have been, or are being, tested in clinical trials involving patients with various types of cancer. Nonetheless, no p53-targeting drug has so far been approved for clinical use. Advances in our understanding of p53 biology provide some clues as to the underlying reasons for the variable clinical activity of p53-restoring therapies seen thus far. In this Review, we discuss the intricate interactions between p53 and its cellular and microenvironmental contexts and factors that can influence p53's activity. We also propose several strategies for improving the clinical efficacy of these agents through the complex perspective of p53 functionality.
Collapse
Affiliation(s)
- Amos Tuval
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm, Sweden
| | | | - Angelos Heldin
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm, Sweden
| | | | - Klas G Wiman
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm, Sweden.
| |
Collapse
|
49
|
Li S, Wang A, Wu Y, He S, Shuai W, Zhao M, Zhu Y, Hu X, Luo Y, Wang G. Targeted therapy for non-small-cell lung cancer: New insights into regulated cell death combined with immunotherapy. Immunol Rev 2024; 321:300-334. [PMID: 37688394 DOI: 10.1111/imr.13274] [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: 09/10/2023]
Abstract
Non-small-cell lung cancer (NSCLC), which has a high rate of metastatic spread and drug resistance, is the most common subtype of lung cancer. Therefore, NSCLC patients have a very poor prognosis and a very low chance of survival. Human cancers are closely linked to regulated cell death (RCD), such as apoptosis, autophagy, ferroptosis, pyroptosis, and necroptosis. Currently, small-molecule compounds targeting various types of RCD have shown potential as anticancer treatments. Moreover, RCD appears to be a specific part of the antitumor immune response; hence, the combination of RCD and immunotherapy might increase the inhibitory effect of therapy on tumor growth. In this review, we summarize small-molecule compounds used for the treatment of NSCLC by focusing on RCD and pharmacological systems. In addition, we describe the current research status of an immunotherapy combined with an RCD-based regimen for NSCLC, providing new ideas for targeting RCD pathways in combination with immunotherapy for patients with NSCLC in the future.
Collapse
Affiliation(s)
- Shutong Li
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Aoxue Wang
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yongya Wu
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Shengyuan He
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Wen Shuai
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Min Zhao
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yumeng Zhu
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Xiuying Hu
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yubin Luo
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Guan Wang
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
Casalin I, De Stefano A, Ceneri E, Cappellini A, Finelli C, Curti A, Paolini S, Parisi S, Zannoni L, Boultwood J, McCubrey JA, Suh PG, Ramazzotti G, Fiume R, Ratti S, Manzoli L, Cocco L, Follo MY. Deciphering signaling pathways in hematopoietic stem cells: the molecular complexity of Myelodysplastic Syndromes (MDS) and leukemic progression. Adv Biol Regul 2024; 91:101014. [PMID: 38242820 DOI: 10.1016/j.jbior.2024.101014] [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: 11/10/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
Myelodysplastic Syndromes, a heterogeneous group of hematological disorders, are characterized by abnormalities in phosphoinositide-dependent signaling, epigenetic regulators, apoptosis, and cytokine interactions within the bone marrow microenvironment, contributing to disease pathogenesis and neoplastic growth. Comprehensive knowledge of these pathways is crucial for the development of innovative therapies that aim to restore normal apoptosis and improve patient outcomes.
Collapse
Affiliation(s)
- Irene Casalin
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy.
| | - Alessia De Stefano
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Eleonora Ceneri
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Alessandra Cappellini
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Carlo Finelli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Antonio Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Stefania Paolini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Sarah Parisi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Letizia Zannoni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Jacqueline Boultwood
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Pann-Ghill Suh
- Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Giulia Ramazzotti
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Roberta Fiume
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Matilde Y Follo
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| |
Collapse
|