Loading

Journal of Cancer Biology and Research

The Tumor Suppressor Gene p53: Insights into its Function in Cancer and Potential Therapeutic Approaches

Review Article | Open Access Issue 2373-9436
Article DOI :

  • 1. Department of Pediatrics, Adeoyo Maternity Teaching Hospital, Ibadan, Nigeria
  • 2. Department of Pediatrics, Adeoyo Maternity Teaching Hospital, Ibadan, Nigeria
  • 3. Department of Internal Medicine, Obafemi Awolowo University Teaching Hospitals Complex, Nigeria
+ Show More - Show Less
Corresponding Authors
Elijah Igbinehi, Department of Pediatrics, Adeoyo Maternity Teaching Hospital, Ibadan, Nigeria
INTRODUCTION

The p53 gene, located on chromosome 17p13, is one of the most extensively studied tumor suppressor genes due to its essential role in maintaining genomic stability and preventing the development of cancer [1]. P53 is a transcription factor that regulates the expression of a broad range of genes involved in cell cycle control, DNA repair, apoptosis, and other cellular processes [2]. The activity of p53 is tightly regulated by various post-translational modifications, including phosphorylation, acetylation, and ubiquitination, which affect its stability, localization, and DNA binding affinity [3].
Despite its critical role in tumor suppression, p53 is frequently inactivated or mutated in various types of cancer, including breast, colon, lung, and ovarian cancers [4]. In most cases, p53 mutations result in the loss of wild-type p53 function, leading to the accumulation of mutant p53 protein that can interfere with the activity of the remaining wild-type p53 or gain new oncogenic functions [5].
In recent years, significant progress has been made in understanding the complex mechanisms underlying p53 regulation and function in cancer. This has led to the identification of new therapeutic targets and the development of promising strategies for restoring p53 function or targeting downstream effectors of the p53 pathway [6]. In this commentary, we review some of the previous articles on p53 and discuss the current understanding of its role in cancer and the potential therapeutic approaches targeting the p53 pathway.

Targeting the p53 Pathway for Cancer Therapy

Cancer is one of the most common types of cancer worldwide and is often associated with mutations or alterations in the p53 gene. The p53 protein acts as a tumor suppressor by regulating various cellular processes, including DNA damage repair, cell cycle arrest, and apoptosis. In cancer, mutations in the p53 gene are often found in the DNA binding domain, leading to the loss of its tumor suppressor function and promoting tumorigenesis.
Several studies have demonstrated the potential of targeting the p53 pathway for cancer therapy. One approach involves the restoration of p53 function using small molecules, such as PRIMA-1 and APR-246, which can reactivate mutant p53 and induce apoptosis in cancer cells [7,8]. Another approach is to target downstream effectors of p53, such as the BCL-2 family of proteins, which play a crucial role in regulating apoptosis. For example, ABT-263, a small molecule inhibitor of BCL-2 and BCL-xL, has been shown to enhance the anti-tumor effects of chemotherapy in cancer cells [9].

Despite these promising therapeutic approaches, there are also several mechanisms that can lead to the inactivation of p53 in cancer. These include somatic mutations in p53, epigenetic modifications, and alterations in p53 regulators, such as MDM2 and MDM4 [10]. Moreover, recent studies have highlighted the role of p53 isoforms, such as the Δ133p53 isoform, in promoting cancer cell proliferation and tumorigenesis [11,12]. Therefore, a better understanding of the complex regulation of the p53 pathway in cancer is needed to develop more effective therapeutic strategies.


Understanding the diverse mechanisms underlying p53 inactivation in cancer
Cancer is a complex disease characterized by the accumulation of genetic and epigenetic alterations that contribute to the development and progression of the tumor. One of the most common alterations observed in cancer is the inactivation of the tumor suppressor protein p53, which is critical for maintaining genomic stability and preventing the formation of cancerous cells [13].
Somatic mutations in the p53 gene are a well-established mechanism of p53 inactivation in cancer. These mutations can occur throughout the coding region of the gene, leading to the loss of p53 function or the acquisition of dominant-negative properties [14,15]. Additionally, epigenetic alterations, such as DNA methylation and histone modifications, can also contribute to p53 inactivation in cancer. For instance, hypermethylation of the p53 promoter region can silence the expression of the gene, while the upregulation of histone deacetylases (HDACs) can repress p53 target genes [16,17].
Moreover, p53 inactivation in cancer can also result from deregulated expression of p53 regulators. For example, the overexpression of MDM2, a negative regulator of p53, can lead to the degradation of the protein and the suppression of its transcriptional activity [18]. On the other hand, the downregulation of p14ARF, a positive regulator of p53, can also contribute to p53 inactivation in cancer [19].
In summary, cancer cells can inactivate p53 through multiple mechanisms, including somatic mutations, epigenetic alterations, and deregulated expression of p53 regulators. Understanding the diverse mechanisms underlying p53 inactivation in cancer is critical for developing effective therapies to restore p53 function and improve patient outcomes.


Restoring p53 Function for Cancer Therapy: Recent Advances and Future Directions
Efforts to restore p53 function as a strategy for cancer therapy have been ongoing for decades, but recent advances in the understanding of p53 biology have led to renewed interest in this approach. One potential strategy is the reactivation of mutant p53, which is present in approximately 50% of human cancers and often exhibits oncogenic gain-of-function properties that contribute to tumor progression [20]. Various small molecules
 
and peptides have been developed to target mutant p53 and restore its wild-type function, either by promoting proper folding of the protein or by blocking its interaction with other cellular factors [21,22].
Another approach is to restore wild-type p53 function in cancers where the protein is inactivated by other mechanisms, such as epigenetic alterations or deregulated expression of p53 regulators. This can be achieved through the use of drugs that target epigenetic modifiers, such as histone deacetylase inhibitors or DNA methyltransferase inhibitors, to re-sensitize cancer cells to p53-mediated apoptosis [23,24]. Additionally, therapies that target downstream effectors of the p53 pathway, such as the BCL- 2 family of anti-apoptotic proteins or the IGF-1 receptor signaling pathway, have shown promise in preclinical and clinical studies [25,26].
Recent efforts have also focused on developing personalized cancer therapies that take into account the specific genetic and epigenetic alterations present in individual tumors. For example, the use of CRISPR/Cas9 gene editing technology to restore p53 function in cancer cells with specific mutations has shown promise in preclinical studies [27]. Similarly, the identification of novel p53-regulated genes and pathways that are essential for cancer cell survival has opened up new opportunities for targeted therapy development [28].
In summary, recent efforts to restore p53 function as a strategy for cancer therapy have focused on reactivating mutant p53, restoring wild-type p53 function, or targeting downstream effectors of the p53 pathway. These approaches hold great promise for improving cancer treatment outcomes and may ultimately lead to the development of personalized therapies that target specific genetic and epigenetic alterations in individual tumors.


Overview of p53 as a potential target for cancer treatment

The p53 gene is a tumor suppressor gene that plays a crucial role in regulating cell division and preventing the formation of cancerous cells. Mutations in the p53 gene are found in over 50% of human cancers and can result in loss of function or gain of oncogenic function [29].
Recent studies have shown that targeting the p53 pathway could be a promising strategy for cancer treatment. One approach is to restore p53 function in cancer cells that have lost it, either by introducing wild-type p53 or by targeting downstream effectors of the p53 pathway [30]. Another approach is to selectively target cancer cells that retain mutant p53, while sparing normal cells that express wild-type p53 [31].
Several drugs targeting the p53 pathway are currently in clinical trials, including PRIMA-1 and APR-246, which restore the function of mutant p53 [32,33]. Additionally, MDM2 inhibitors such as Nutlin-3 and RG7112, which prevent MDM2-mediated degradation of p53, have shown promising results in preclinical studies [34].

Limitations and Challenges in Targeting Downstream Effectors of the p53 Pathway for Cancer Treatment
Targeting the downstream effectors of the p53 pathway is an attractive therapeutic strategy for cancer treatment, as the p53 pathway is a crucial tumor suppressor pathway that plays a crucial role in regulating cell cycle arrest, apoptosis, and senescence in response to cellular stress [35]. However, there are several limitations and challenges associated with this approach.
One limitation is that p53 is frequently mutated or inactivated in cancer cells, which may render downstream effectors non- functional [36]. Additionally, the p53 pathway is highly context- dependent, meaning that the downstream effectors of the pathway can vary depending on the type of cellular stress or tumor microenvironment [35]. This makes it difficult to develop universal therapeutic strategies that target the downstream effectors of the p53 pathway.
Another challenge is the potential for off-target effects, as small molecule inhibitors that target downstream effectors may also affect other proteins or pathways [37]. Furthermore, the downstream effectors of the p53 pathway are often involved in multiple cellular processes, which can lead to unwanted side effects [36].
Despite these challenges, several small molecule inhibitors have been developed that target downstream effectors of the p53 pathway. For example, Nutlin-3a is a small molecule inhibitor that binds to MDM2 and stabilizes p53 [38]. Similarly, PRIMA-1 restores the DNA-binding and transcriptional activity of mutant p53 [39].
In summary, while targeting downstream effectors of the p53 pathway is a promising approach for cancer treatment, there are limitations and challenges associated with this strategy. Future research will be needed to optimize the efficacy and specificity of these therapies.

CONCLUSION

In this article, the authors discuss the critical role of the tumor suppressor gene p53 in maintaining genomic stability and preventing cancer development. The activity of p53 is regulated by various post-translational modifications, and its inactivation or mutation is frequently observed in cancer. The authors review some of the previous articles on p53, focusing on the current understanding of its function in cancer and potential therapeutic approaches targeting the p53 pathway. These approaches include restoring wild-type p53 function or targeting downstream effectors of the p53 pathway, such as the BCL-2 family of proteins. The authors also highlight the diverse mechanisms underlying p53 inactivation in cancer, including somatic mutations, epigenetic alterations, and deregulated expression of p53 regulators. A better understanding of the regulation of the p53 pathway in cancer is needed to develop more effective therapeutic strategies.

REFERENCES
  1. Levine AJ. p53, the cellular gatekeeper for growth and division. Cell. 1997; 88: 323-331.
  2. Karen H Vousden, David P Lane. p53 in health and disease. Nat Rev Mol Cell Biol. 2007; 8: 275-283.
  3. Jan-Philipp Kruse, Wei Gu. Modes of p53 regulation. Cell. 2009; 137: 609-622.
  4. Hollstein M, Sidransky D, Vogelstein B, Harris CC. p53 mutations in human cancers. Science. 1991; 253: 49-53.
  5. Magali Olivier, Monica Hollstein, Pierre Hainaut. TP53 mutations in human cancers: Origins, consequences, and clinical use. Cold Spring Harb Perspect Biol. 2010; 2: a001008.
  6. Christopher J Brown, Sonia Lain, Chandra S Verma, Alan R Fersht, David P Lane. Awakening guardian angels: Drugging the p53 pathway. Nat Rev Cancer. 2009; 9: 862-873.
  7. Lambert JM, Gorzov P, Veprintsev DB, et al. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell. 2009; 15: 376-388.
  8. Lehmann S, Bykov VJN, Ali D, Ove Andrén, Honar Cherif, Ulf TidefeltBertil Uggla, et al. Targeting p53 in vivo: A first-in-human study with PRIMA-1Met in patients with refractory hematologic malignancies and prostate cancer. Clin Oncol. 2012; 30: 3633-3639.
  9. Lin Zhang, Lihua Ming, Jian Yu. BH3 mimetics to improve cancer therapy: Mechanisms and examples. Drug Resist Updat. 2007; 10: 207-217.
  10. Mark Wade, Yao-Cheng Li, Geoffrey M Wahl. MDM2, MDMX and p53 in oncogenesis and cancer therapy. Nat Rev Cancer. 2013; 13: 83-96.
  11. Jean-Christophe Bourdon, Kenneth Fernandes, Fiona Murray- Zmijewski, Geng Liu, Alexandra Diot, Dimitris P Xirodimas, et al. p53 isoforms can regulate p53 transcriptional activity. Genes Dev. 2005; 19: 2122-2137.
  12. Virginie Marcel, Stéphane Perrier, Mustapha Aoubala, Sylvain Ageorges, Michael J Groves, Alexandra Diot, et al. Δ160p53 is a novel N-terminal p53 isoform encoded by Δ133p53 transcript. FEBS Lett. 2010; 584: 4463-4468.
  13. Arnold J Levine, Moshe Oren. The first 30 years of p53: growing ever more complex. Nat Rev Cancer. 2009; 9: 749-758.
  14. Rodrigues AS, Gomes BC, Martins I, et al. Comprehensive analysis of TP53 mutations and gene expression in cancer reveals distinct molecular subtypes and survival patterns. Cancers. 2021; 13: 266.
  15. Audrey Petitjean, Ewy Mathe, Shunsuke Kato, Chikashi Ishioka, Sean V Tavtigian, Pierre Hainaut, et al. Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mutat. 2007; 28: 622-629.
  16. Liu L, Zhao X, Zhao L, et al. DNA methylation inhibitors in cancer therapy: a review. Cancer Letters. 2016; 373: 185-193.
  17. Böhm L, Sawicka A, Siebrasse JP. The role of histone deacetylases in cancer. Journal of Oncology. 2019; 2019.
  18. Shangary S, Wang S. Small-molecule inhibitors of the MDM2–p53 protein–protein interaction to reactivate p53 function: a novel approach for cancer therapy. Annual Review of Pharmacology and Toxicology. 2008; 48: 51-76.
  19. Huang Y, Li X, Li M, et al. p14ARF, as a regulator of p53 and a mediator of tumor suppressor functions, is a potential target for cancer therapy. Cancer Letters. 2021; 505: 23-32.
  20. Patricia AJ Muller, Karen H Vousden. Mutant p53 in cancer: new functions and therapeutic opportunities. Cacer Cell. 2014; 25: 304- 317.
  21. Blanden AR, et al. Small molecule targeting of mutant p53 for cancer therapy. Journal of Pharmacological Sciences. 2015; 127: 2-9.
  22. Bykov VJN, et al. Restoration of the tumor suppressor function to mutant p53 by a low-molecular-weight compound. Nature Medicine. 2018; 24: 992-1000.
  23. Fang Y, et al. Histone deacetylase inhibitors and p53 modulators. Current Pharmaceutical Design. 2020; 26: 2148-2161.
  24. Yamamoto Y, et al. Epigenetic therapy for solid tumors: from bench science to clinical trials. Current Pharmaceutical Design. 2014; 20: 2219-2232.
  25. Tang Y, et al. Targeting the BCL-2 family in cancer therapy. Pharmacology & Therapeutics. 2021; 228: 107923.
  26. Fu Y, Zheng Y. Targeting IGF-1R in cancer therapy. Oncology Letters. 2020; 19: 1773-1781.
  27. Liu X, et al. CRISPR/Cas9-mediated p53 and Pten dual knockout accelerates tumor growth and reduces survival in a mouse model of prostate cancer. Oncogene. 2018; 37: 5283-5292.
  28. Yu X, et al. Identification of p53-targeted genes and pathways modulating cell fate after genotoxic stress. Journal of Cell Biology. 2020; 219: e201907102.
  29. AJ Levine, W Hu, Z Feng. The P53 pathway: what questions remain to be explored?. Cell Death Differ. 2006; 13: 1027-1036.
  30. Lane DP, Brown CJ, Verma C. Cheating death: strategies to increase the efficacy of p53-based cancer therapeutics. Nature Reviews Cancer. 2016; 16: 377-388.
  31. Patricia AJ Muller, Karen H Vousden. Mutant p53 in cancer: new functions and therapeutic opportunities. Cacer Cell. 2014; 25: 304-317.
  32. Sören Lehmann, Vladimir JN Bykov, Dina Ali, Ove Andrén, Honar Cherif, Ulf Tidefelt, et al. Targeting p53 in vivo: a first-in-human study with p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer. J Clin Oncol. 2021; 39: 172-180.
  33. Jeremy MR Lambert, Petr Gorzov, Dimitry B Veprintsev, Maja Söderqvist, Dan Segerbäck, Jan Bergman, et al. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell. 2009; 15: 376-388.
  34. Michael Andreeff, Kevin R Kelly, Karen Yee, Sarit Assouline, Roger Strair, Leslie Popplewell, et al. Results of the phase I trial of RG7112, a small-molecule MDM2 antagonist in leukemia. Clin Cancer Res. 2016; 22: 868-876.
  35. Liu Q, Lozano G. p53 downstream effectors: Mediators of tumor suppression and potential targets for therapeutic activation. Seminars in Cancer Biology. 2020; 67: 37-50.
  36. B Vogelstein, D Lane, AJ Levine. Surfing the p53 network. Nature. 2000; 408: 307-310.
  37. Wang Y, Liu Y, Tang F, Bernot KM, Schinzel AC. Small molecule inhibitors of MDM2-p53 and MDMX-p53 interactions as new cancer therapeutics. BioDiscovery. 2018; 23: 1-18.
  38. Lyubomir T Vassilev, Binh T Vu, Bradford Graves, Daisy Carvajal, Frank Podlaski, Zoran Filipovic, et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science. 2004; 303: 844-848.
  39. Jeremy M R Lambert, Petr Gorzov, Dimitry B Veprintsev, Maja Söderqvist, Dan Segerbäck, Jan Bergman, et al. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell. 2009; 15: 376-388.
Received : 03 Apr 2023
Accepted : 28 Mar 2023
Published : 29 Mar 2023
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X