Loading

Journal of Immunology and Clinical Research

PD-L1, Inflammation and Glioblastoma

Review Article | Open Access

  • 1. Neuroinflammation Unit, Biotech research and innovation center (BRIC), Copenhagen University, Copenhagen, Denmark
+ Show More - Show Less
Corresponding Authors
Robert Carlsson, Neuroinflammation Unit, Biotech and research innovation center (BRIC), Copenhagen University, Copenhagen, Denmark
CONCLUDING REMARKS

Given the heterogeneity and complex etiology of the GBM and the origin of the tumor cells, it is not surprising that the environment that surrounds the tumors display wide varieties of responses. In particular, the immunosuppressive molecule PD-L1 has heterogenous functions depending on the context of expression. Hence, PD-L1 can convey immunosuppressive properties; 1. by inhibiting cytotoxic T-lymphocytes from destroying the tumor cells, 2. by contributing to sustained chronic inflammation as a result of modulating the immune cells so that the tumor microenvironment is activated by invasion of immune suppressive regulatory T cells, and/or by mediating the release of inflammatory mediators by immune cells with no anti-tumor activity. In some cancers, PD-L1 expression by tumor cells correlates with a good prognosis for the patient, whereas in glioblastoma many reports suggest that it is a bad prognostic marker. The interplay between the gliomas, the tumor microenvironment and infiltrating immune cells can hold key information to how to utilize PD-L1/PD-1 directed immunotherapies to successfully treat GBM patients. We suggest that the PD-L1 pathway can exert positive and negative effects on the GBMs (Figure 1). This divergent effect might be dependent on the glioma subclass, expression levels of PD-L1 and PD-L1 regulatory molecules, but most importantly on the cell type that express PD-L1 in the tumor microenvironment.

REFERENCES

1. Chen R, Jiang X, Sun D, Han G, Wang F, Ye M, et al. Glycoproteomics analysis of human liver tissue by combination of multiple enzyme digestion and hydrazide chemistry. J Proteome Res. 2009; 8: 651-661.

2. Dong H, Zhu G, Tamada K, Chen L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med. 1999; 5: 1365-1369.

3. Liu Y, Carlsson R, Ambjørn M, Hasan M, Badn W, Darabi A, et al. PD-L1 expression by neurons nearby tumors indicates better prognosis in glioblastoma patients. J Neurosci. 2013; 33: 14231-14245.

4. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002; 8: 793-800.

5. Parsa AT, Waldron JS, Panner A, Crane CA, Parney IF, Barry JJ, et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2007; 13: 84-88.

6. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 2007; 27: 111-122.

7. Paterson AM, Brown KE, Keir ME, Vanguri VK, Riella LV, Chandraker A, et al. The programmed death-1 ligand 1:B7-1 pathway restrains diabetogenic effector T cells in vivo. J Immunol. 2011; 187: 1097-1105.

8. Cheng X, Veverka V, Radhakrishnan A, Waters LC, Muskett FW, Morgan SH, et al. Structure and interactions of the human programmed cell death 1 receptor. J Biol Chem. 2013; 288: 11771-11785.

9. Akbay EA, Koyama S, Carretero J, Altabef A, Tchaicha JH, Christensen CL, et al. Activation of the PD-1 Pathway Contributes to Immune Escape in EGFR-Driven Lung Tumors. Cancer Discov. 2013; 3: 1355- 1363.

10. Wintterle S, Schreiner B, Mitsdoerffer M, Schneider D, Chen L, Meyermann R, et al. Expression of the B7-related molecule B7-H1 by glioma cells: a potential mechanism of immune paralysis. Cancer Res. 2003; 63: 7462-7467.

11. Spranger S, Spaapen RM, Zha Y, Williams J, Meng Y, Ha TT, et al. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells. Sci Transl Med. 2013; 5: 200ra116.

12. Bloch O, Crane CA, Kaur R, Safaee M, Rutkowski MJ, Parsa AT. Gliomas promote immunosuppression through induction of B7-H1 expression in tumor-associated macrophages. Clin Cancer Res. 2013; 19: 3165- 3175.

13. Hatten ME, Shelanski ML. Mouse cerebellar granule neurons arrest the proliferation of human and rodent astrocytoma cells in vitro. J Neurosci. 1988; 8: 1447-1453.

14. Kingwell K. Neuro-oncology: Glioblastoma prognosis linked to neuronal PD-L1 expression in tumour-adjacent tissue. Nat Rev Neurol. 2013; 9: 602-603.

15. Issazadeh-Navikas S. Alerting the immune system via stromal cells is central to the prevention of tumor growth. OncoImmunology. 2013; 2: e27091.

16. Friedmann-Morvinski D, Bushong EA, Ke E, Soda Y, Marumoto T, Singer O, et al. Dedifferentiation of neurons and astrocytes by oncogenes can induce gliomas in mice. Science. 2012; 338: 1080-1084.

17. Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell. 2006; 9: 157-173.

18. Lee J, Kotliarova S, Kotliarov Y, Li A, Su Q, Donin NM, et al. Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell. 2006; 9: 391-403.

19. Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, et al. The somatic genomic landscape of glioblastoma. Cell. 2013; 155: 462-477.

20. Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010; 17: 98-110.

21. Zhu H, Acquaviva J, Ramachandran P, Boskovitz A, Woolfenden S, Pfannl R, et al. Oncogenic EGFR signaling cooperates with loss of tumor suppressor gene functions in gliomagenesis. Proc Natl Acad Sci U S A. 2009; 106: 2712-2716.

22. Doucette T, Rao G, Rao A, Shen L, Aldape K, Wei J, et al. Immune Heterogeneity of Glioblastoma Subtypes: Extrapolation from the Cancer Genome Atlas. Cancer Immunology Research. 2013; 1: 112- 122.

23. Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. Inflammationinduced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer. 2013; 13: 759-771.

24. Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 2012; 4: 127ra37.

25. Bos PD, Plitas G, Rudra D, Lee SY, Rudensky AY. Transient regulatory T cell ablation deters oncogene-driven breast cancer and enhances radiotherapy. J Exp Med. 2013; 210: 2435-2466.

26. Sielska M, Przanowski P, Wylot B, Gabrusiewicz K, Maleszewska M, Kijewska M, et al. Distinct roles of CSF family cytokines in macrophage infiltration and activation in glioma progression and injury response. J Pathol. 2013; 230: 310-321.

27. Söderberg-Nauclér C, Rahbar A, Stragliotto G. Survival in patients with glioblastoma receiving valganciclovir. N Engl J Med. 2013; 369: 985- 986.

28. Bhat KP, Balasubramaniyan V, Vaillant B, Ezhilarasan R, Hummelink K, Hollingsworth F, et al. Mesenchymal differentiation mediated by NFkB promotes radiation resistance in glioblastoma. Cancer Cell. 2013; 24: 331-346.

29. Ebner K, Bandion A, Binder BR, de Martin R, Schmid JA. GMCSF activates NF-kappaB via direct interaction of the GMCSF receptor with IkappaB kinase beta. Blood. 2003; 102: 192-199.

30. Huang G, Wen Q, Zhao Y, Gao Q, Bai Y. NF-kB plays a key role in inducing CD274 expression in human monocytes after lipopolysaccharide treatment. PLoS One. 2013; 8: e61602.

31. Kanai T, Totsuka T, Uraushihara K, Makita S, Nakamura T, Koganei K, et al. Blockade of B7-H1 suppresses the development of chronic intestinal inflammation. J Immunol. 2003; 171: 4156-4163.

32. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012; 366: 2455-2465.

33. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012; 366: 2443-2454.

34. Latchman YE, Liang SC, Wu Y, Chernova T, Sobel RA, Klemm M, et al. PD-L1-deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells. Proc Natl Acad Sci U S A. 2004; 101: 10691-10696

Abstract

Programmed death ligand 1 (PD-L1) is a transmembrane receptor that can contribute both to the termination of unwanted chronic inflammation, and to immune escape from desired immune responses by glioblastoma multiforme (GBM). Recent reports show that expression of the anti-inflammatory protein, PD-L1 on glioblastoma and other cancer cells, correlate with poor prognosis as a result of immune escape. On the contrary on stromal cells such as neurons, PD-L1 expression can correlate with a better prognosis, suggesting that the cell type where PD-L1 is expressed plays decisive role for the outcome of the disease. We discuss here the divergent functions of PD-L1 in the tumor microenvironment.

Keywords


• Tumor microenvironment
• Stromal cell function
• EGFR PD-L1
• Glioblastoma
• Inflammation
 

Citation

Carlsson R, Issazadeh-Navikas S (2014) PD-L1, Inflammation and Glioblastoma. J Immunol Clin Res 2(1): 1013.

INTRODUCTION

Glioblastoma multiforme (GBM) is a grade IV WHO tumor with dismal patient prognosis and a median survival time of 12- 14 months. Despite surgery and cytostatic treatment the tumor prevails. Better understanding of the initiation, maintenance, microenvironment and etiology of GBM is required for designing more successful treatments.

PD-L1 and GBM

Programmed death ligand 1 (PD-L1, also known as CD274 and B7H1) is a trans-membrane protein molecule with suggested proteoglycan moieties [1]. It is expressed predominantly in the heart and placenta at the mRNA level [2]. At the protein level PD-L1 expression can be found and induced on macrophages, T-lymphocytes and neurons [3].

The PD-L1 protein has an extracellular domain, a transmembrane domain and an intracellular domain. The extracellular domain (ECD) of the PD-L1 protein is comprised of IgC–like and IgV-like domains [2]. The ECD of PD-L1 interacts with the programmed death 1 (PD-1, also known as PDCD1) and B7.1 receptors. Blocking of the PD-L1 molecule with antibodies blocks cancer cells from killing cytotoxic T lymphocytes (CTLs), whereas siRNA knockdown of the PD-L1 molecule in glioblastoma makes the glioblastoma susceptible to CTL mediated killing [4,5]. Interaction with B7.1 results in the inhibition of T-lymphocyte responses in vitro and diabetogenic T-lymphocytes in vivo [6, 7].

PD-L1: PD-1 has an affinity of 0.77uM when glycosylated and in dimers and the monomeric non-glycosylated proteins has an affinity of 26.6uM. Dimeric PD-L1:B7.1 bind with an affinity of 1.99uM and the non-glycosylated monomeric proteins 80.7uM[6,8]. This shows that important receptor ligand interactions can be made even though the apparent affinity is relatively low [8].

Of note, no functional properties are known to date for the intracellular domain of PD-L1, although some indications of an outside-in signaling exist upon B7.1 binding. CTLA4 and CD28 are the two other ligands for B7.1. When co-culturing CTLA4-/- CD28-/- T-lymphocytes with beads coupled with CD3 and B7.1 their proliferation is diminished, suggesting that B7.1 on the beads binds to PD-L1 on the T lymphocytes and down modulate anti-CD3 provoked proliferative capacity [6].

PD-L1 has been suggested to promote immune escape by tumors including glioblastomas [4,9,10]. Loss of phosphatase and tensin homolog (PTEN), a major GBM tumor suppressor, induces the expression of PD-L1 and possibly immune escape in GBM [5]. Impact of PTEN loss on up-regulation of PD-L1 in PTEN-GBMs is not the result of the gene regulation but it regulates PD-L1 post transcriptionally and correlates with the induction of PI3K. In melanoma the expression of PD-L1 is induced by infiltration of CD8+ T-lymphocytes [11], but a similar mechanism is not reported in GBM. Macrophages from GBM patients express higher levels of PD-L1 in the peripheral blood and in the tumor tissues [12]. Collectively these studies of immune evasion and the mechanism of escape would be for glioblastoma cells to express the PD-L1 protein that kills PD-1/B7.1 expressing T-lymphocytes invading the tumor. Except for immune evasion, it is yet unknown if PD-L1 expression by tumor cells serve additional biological advantages for malignant tumors to establish, grow and invade. Studies in vitro on co-cultures of cerebellar granular neurons (CGNs) and astrocytoma cells have shown that CGNs can inhibit the proliferation of the astrocytoma cells [13]. We have shown that killing of murine glioblastoma (GL261) cells is dependent on the expression and activity of PD-L1 receptor on neurons [3,14,15]. Furthermore, the killing of the gliomas was accompanied by upregulation of cleaved Caspase-3 and other hall markers of apoptosis [3]. This is the first finding describing the mechanism of action by which neurons kill glioblastoma cells.

The differential effect of expression of PD-L1 on neurons and on glioblastoma raises a question; is the PD-L1 on neurons different from the PD-L1 expressed on the glioblastoma? Given that the PD-L1 protein can be glycosylated differentially, and/or bound to differential binding partners depending on the cellular context, one could speculate that there are either differences in the PD-L1 molecule itself, or among proteins it interacts with, that makes the difference between the neurons and the glioblastoma. An additional explanation might be that the glioblastomas expressing the PD-L1 molecule have disarmed it, by either e.g. differential glycosylation or proteins interaction, so that it is not conferring killing of nearby glioblastoma cells.

Glioblastoma subtypes

GBM cells within a tumor are morphologically and transcriptionally diverse. Investigation of the impact of silencing the oncosuppressors neurofibromatosis type I (NF1) and protein 53 (P53) for transformation of primary cells suggests that GBM can arise from many different types of cells, ranging from neural stem cells to astrocytes. In fact adult neurons were elegantly shown to be the origin of tumors for some of the evoked GBMs [16].

Despite the heterogeneity, molecular subtypes have been characterized; poor prognosis, as determined by genetic profiling, includes mesenchymal GBMs and tumors expressing neural stem cell markers, while tumors tumors expressing a proneural profile correlated with better prognosis [17]. Interestingly, only special culture conditions in vitro, i.e. addition of Epidermal growth factor (EGF) and Fibroblast growth factor (FGF) and no serum, maintain the in vivo gene expression profiles [18]. Classic glioblastomas often have amplifications or activating mutations in the Epidermal growth factor receptor (EGFR) gene resulting in overexpression and phosphorylation of the EGFR protein, whereas the oncosuppressor P53 rarely is mutated [19,20].

EGFR and Glioblastoma

EGFR mutation is one of the main driver mutations in GBM. EGFR is a transmembrane receptor tyrosine kinase (RTK) oncogene. Upon binding to EGF, the signaling pathway is activated and thereby it induces cell proliferation and/or differentiation. Alteration of EGFR activity can be triggered by mutations, fusion to nearby genes, inversions or amplification of the egfr gene [19]. These alterations of the egfr gene are present in more than 40% of GBMs. Normally the activation of EGFR requires EGF ligand binding, but in some occasions EGFR is mutated to escape requirement of EGF. EGFRvIII is a truncated version of the EGFR receptor that signals constitutively without the need for EGF. The frequency of the EGFRvIII mutation in GBM is debated but recent technology advances have shown it to be highly expressed in 11% GBM [19]. Recently the PD-1 pathway and EGFR were correlated and shown to be simultaneously expressed in a lung cancer model [9]. EGFR activity enhances the expression levels of the PD-L1 protein in vitro, as judged by the loss of cell surface staining of PD-L1 by lung cancer cells after treatment with an EGFR inhibitor, Gefitinib. Transformation of the bronchial epithelial cell line with a EGFR mutant enhances the expression of PD-L1 both at the mRNA level and as a protein on the cell surface. This study points out immune escape and a reduction in immune responses as the reason for the coexpression of EGFR and PD-L1 on the cancer cells [9]. Collectively these data suggest that the GBMs over expressing the EGFR molecule might also overexpress the PD-L1 molecule. In a mouse model of GBM it was shown that the EGFR tumorigenic activity is enhanced by deletion of PTEN. Loss of PTEN is inducing the expression of PD-L1 suggesting a link between over expression of EGFR, loss of PTEN and subsequent gain of PD-L1 expression [21]. Among the EGFR and PD-L1 molecules it is rather PD-L1 that seems to correlate with severity in GBM, as it is highly expressed in mesenchymal GBMs, suggesting the importance for PD-L1, the immune system and inflammation in GBM disease [22].

Inflammation and GBM

Inflammation can be both friend and foe in cancer. For certain cancer subtypes such as colon cancer, inflammation is suggested to be an inducer of mutations driving cancer progression together with the gut microbiota [23]. It is important to differ between chronic inflammation (CI) and a desired inflammation (DI), which is a physiological immune response fighting the tumor. For DI it has been suggested that PD-L1 is upregulated on tumors as a response to the immune attack and PD-L1 expression confers a better cancer prognosis in patients [24]. Conversely, PD-L1 expression on GBM has been shown to correlate with a bad prognosis [3]. Those in vivo findings are in contrast to each other and suggest an opposing outcome of PD-L1 expression on cancer cells depending on the cancer type.

A direct link between immune suppression, CI and the severity of disease was recently shown in a cancer model in vivo. Depletion of suppressive T regulatory cells reduces tumor burden and CI in a breast cancer animal model [25]. For GBMs similar role for inflammation has been suggested. Immune surveillance through DI might be important to constrain the growth of the tumor cells but CI itself is most likely a negative factor. Cancer cells could also contribute to induction and/or maintenance of CI which in turn nourishes tumor development. In a murine GL261- induced glioma model, it is shown that gliomas can produce granulocyte macrophage colony-stimulating factor (GM-CSF) and stimulate infiltrating macrophages and microglia, suggesting that the malignant cells help maintain a CI state of the tumor [26].

Cytomegalovirus (CMV) DNA and/or protein products are often expressed in GBM. Valganciclovir (an antiviral drug) was used to treat the GBM in combination with surgery. Patients treated with the antiviral drug had an increased survival time (50%) compared to patients treated with surgery only [27], suggesting that a chronic CMV infection could possibly contribute to CI and thereby to GBM progression. Further more, nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB) has been shown to induce proneural to mesenchymal transition of GBMs and correlated with CD44 expression [28]. Xenografts of mesechymal GBMs into immune compromised mice converted the GBMs from the mesenchymal subtype to the proneural, suggesting that an inflammatory condition is necessary to maintain the mesenchymal GBM phenotype [28]. Tumor necrosis factor alpha (TNF-α) could transform proneural to mesenchymal GBMs via the activity of NFkB. In addition the activity of NFkB regulated the expression of signal transducer and activator of transcription 3 (STAT3), CCAAT/enhancer-binding protein alpha (C/EBP-α) and transcriptional coactivator with PDZ-binding motif (TAZ) known to induce the mesenchymal GBM subtype [28]. Mesenchymal GBMs are among the most aggressive of GBMs expressing high amounts of PD-L1 [22]. In fact, NFkB has been shown to bind and up regulate the GM-CSF promoter in endothelial cells [29], and the PD-L1 promoter in macrophages in inflammatory conditions [30]. In addition, PD-L1 blockade has been shown to inhibit inflammation in a model of CI, suggesting a role for PD-L1 in maintaining the CI state [31]. Although a direct link needs to be established, NFkB induced expression of PD-L1 might be one of the inducers of the CI observed in GBM.

The molecular profile with low expression of PTEN and over expression of PD-L1, NFkB, GM-CSF and EGFR suggests that high grade GBMs, in addition to upregulating molecules that convey proliferative signals, express molecules that induces and maintains CI in the microenvironment of the tumor. In Figure 1 we schematize concerning the issues around PD-L1 expression in CI (A) and on neurons (B) as an example of the diverse affects PD-L1 expression can have within the tumor microenvironment.

PD-L1 directed immunotherapy in cancer

Two monoclonal antibodies are now available for treatment of the PD-1/PD-L1 axis. Both antibodies have shown to be efficacious in some solid tumor cancers [32,33].

For the PD-1 antibody therapy was efficient in 36% of the patients when PD-L1 was expressed on the tumor [33]. PD-L1 blockage by antibody induced tumor regression with an objective response rate of 6-17 % and prolonged stabilization of disease (12-41%) at 24 weeks [32]. Though the mechanism of action of these antibodies is debated, it is suggested by some that they act through the inhibition of immune escape [32, 33], but there is a possibility that the antibodies increase systemic levels of T-lymphocytes or both inhibit immune escape and increase levels of T-lymphocytes. Activation of anti-tumor T-lymphocytes is desirable to fight cancer progression. Therefore, it is of note to mention that activated T-lymphocytes are reported to express PD-L1 on their surface [34]. To our knowledge the direct importance of PD-L1 expressing T-lymphocytes for their anti-tumor activity has not been addressed. What an eventual anti-PD-L1 immunotherapy could impact suppressing desirable activated T-lymphocytes is also a relevant issue to consider.

Despite the fact that the mechanism of action for PD-L1/PD-1 axis therapy in cancer is still discussed, it might be of importance to carefully consider when one should target the PD-L1/PD-1 axis (anti-PD-L1 in particular) in GBM, since there is a risk for sustained chronic inflammation and GBM progression upon treatment. This is concomitant with our results from studying GBM patients and its model, where high expression of PD-L1 on neurons adjacent to the gliomas correlated with better clinical prognosis likely because neuronal PD-L1 induces apoptotic cell death of gliomas [3]. Treatment with anti-PD-L1 in such setting could preclude neuronal killing activity and hence glioma progression. A similar scenario could be involved in other types of cancer where PD-L1 expression on stromal cells could contribute to prevention of tumor growth.

Received : 13 Dec 2013
Accepted : 15 Jan 2014
Published : 20 Jan 2014
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
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 Cancer Biology and Research
ISSN : 2373-9436
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