Loading

Journal of Cancer Biology and Research

FOXM1: A Promising Target for Cancer Stemness

Review Article | Open Access | Volume 11 | Issue 2

  • 1. Department of Nanomedicine, Houston Methodist Research Institute, USA
  • 2. Department of Stem Cell, Ege University, Turkey
+ Show More - Show Less
Corresponding Authors
Bulent Ozpolat, Department of Nanomedicine, Houston Methodist Research Institute, USA
Abstract

FOXM1 is an oncogenic transcription factor that promotes tumor growth, metastasis, and progression of drug resistance, and its high expression correlates with poor patient survival across various solid tumors. Mutations in the p53 tumor suppressor, a common genetic alteration in approximately half of the human tumors, lead to FOXM1 overexpression. Emerging evidence indicates that a small subpopulation of cancer cells, cancer stem cells (CSCs), within tumors contributes to relapse, metastasis, and progression due to their roles in tumor initiation, self-renewal, differentiation, and therapy resistance. The clinical significance of CSCs on patient prognosis underscores the need for CSC-targeted therapies. FOXM1 plays a significant role in CSC-proliferation and survival, interacting with key stemness-related and pluripotency-associated transcription factors such as SOX2, OCT4, NANOG, and stem cell markers CD133, CD44, CD24, and ALDH. Notably, in-vivo genetic targeting of FOXM1 has been shown to block tumor growth in experimental models. However, there is currently no FDAapproved therapy specifically targeting FOXM1. Given its pivotal role, developing FOXM1-targeted therapies holds significant potential to overcome therapy resistance, halt cancer progression, and improve patient survival. This review evaluates the latest data on the role of FOXM1 in CSCs and explores potential strategies for targeting FOXM1 as a therapeutic approach.

Keywords

• FOXM1

• Cancer stem cells

• Drug resistance

• FOXM1 inhibitors

• Stemness

CITATION

Biltekin E, Ozpolat B (2024) FOXM1: A Promising Target for Cancer Stemness. J Cancer Biol Res 11(2): 1146.

ABBREVIATIONS

ABC Transporter: ATP Binding Cassette Transporter; AFP: Alfa-fetoprotein; BC: Breast Cancer; CC: Cervical Cancer; CSCs: Cancer Stem Cells; EMT: Epithelial-Mesenchymal Transition; FOX: Forkhead-box; GBM: Glioblastoma; HCC: Hepatocellular Carcinoma; LCSCs: Liver Cancer Stem Cells; NPC: Nasopharyngeal Cancer; OCSCs: Ovarian Cancer Stem Cells; TNBC: Triple Negative Breast Cancer; Ref.: References.

INTRODUCTION

In 2024, over 2 million new cancer cases are expected in the United States alone, while globally, cancer remains the second leading cause of death [1,2]. Despite significant advancements in cancer treatments, such as immunotherapy, targeted therapies, and nanomedicine, resistance to these new treatments, as well as conventional therapies like chemotherapy and radiotherapy, remains a critical challenge. This resistance contributes to relapses and poor patient survival [3-5].

CANCER STEM CELLS AND TUMORIGENESIS

Oncogenic transition from healthy cells is driven by various intrinsic and extrinsic factors. Extrinsic factors such as inflammation, viral infections, chemical exposure, radiation, unhealthy dietary habits, and ageing can facilitate oncogenesis. These factors lead to genetic mutations and epigenetic alterations, which are considered intrinsic mediators, alongside heredity factors [6,7].

Cancer stem cells (CSCs), a small but distinct population within tumors, play a crucial role in tumor initiation, metastasis, and relapses. CSCs are characterized by their ability to self-renew, differentiate, proliferate, produce progenitor cells, and resist treatments [4,8-12]. The concept of CSCs was first established in acute myeloid leukemia with CD34+/CD38- leukemia initiating cells. Subsequent research on solid tumors demonstrated that CD44+/CD24-/Lin- cells could form tumors in severe immune- deficient mice with breast cancer [11-13].

To identify CSCs and understand intratumoral heterogeneity, various cell surface markers such as CD44, CD24, C133, CD34, CD38, CD133, CD117, CD271, CXCR4, Lgr5, EPCAM, and ALDH enzyme activity have been widely used [10,14-20]. For instance, Leng et al. showed that LGR5+/CD44+/EPCAM+ cells had a higher tumor-initiating capacity and could form spheroids, unlike CD44-/EPCAM- cells, with Lgr5 positivity playing a major role [10]. Similarly, Fumagalli et al. highlighted the metastatic abilities and plasticity of CSCs, showing that LGR5- cells could migrate and extravasate into blood circulation, but expressed LGR5 upon liver inoculation, whereby ablation of LGR5 expression inhibits the liver metastasis of inoculated LGR5– cells [9]. In glioblastoma model, treatment with the chemotherapeutic agent Temozolamide increased the number of CSCs by converting non-CSCs to CSCs [21]. The dormancy, high expression of pluripotency-related transcription factors, and high drug efflux capacity of CSCs contribute to their drug resistance [22,23].

FOXM1: AN EMERGING MOLECULAR TARGET FOR CSCS

FOXM1 is a transcription factor belonging to the forkhead-box (FOX) protein family, which comprises 19 different subclasses characterized by a “winged-helix” structure [24,25]. FOX proteins play essential roles in development, cell differentiation, cellular hemostasis, apoptosis, and tumorigenesis [25-28]. FOXM1, which has four subtypes (FOXM1a, FOXM1b, FOXM1c, FOXM1d), is involved in embryogenesis, neuronal stem cell proliferation, lung vasculogenesis, and hepatoblastoma proliferation [29-32]. Dysregulation of FOXM1 has been observed in various cancers and is linked to cancer cell proliferation, survival, epithelial- mesenchymal transition (EMT), invasion, migration, poor patient survival, treatment resistance, and cancer stemness [33-35]. FOXM1 is suppressed by the tumor suppressor transcription factor p53, and mutations in p53, present in a half of human tumors, lead to overexpression of FOXM1 [36,37] [Figure 1].

FOXM1, A Promising Target for Cancer Stemness

Figure 1: FOXM1, A Promising Target for Cancer Stemness

FOXM1 IS LINKED TO POOR PROGNOSIS AND SHORTER PATIENT SURVIVAL

FOXM1 is overexpressed in a broad range of tumors, and its upregulation is associated with advanced tumor stages. A meta- analysis by Li et al. revealed that more than half of the patients with solid tumors exhibited higher FOXM1 protein expression, correlating with poor overall survival [38]. Analysis of the TCGA database showed that 22 out of 31 different tumor types have FOXM1 overexpression; in addition, patients with metastatic tumors express significantly higher levels of FOXM1 compared with early-stage cancer [39]. Our recent Kaplan Meier Plotter Survival Analysis showed that higher FOXM1 mRNA expression is significantly correlated with poor patient survival in breast, lung, pancreatic, ovarian, and metastatic gastric cancers [Figure 2].

Kaplan Meier Survival Analysis of FOXM1 mRNA Expression on Overall Survival of Breast Cancer, Lung Cancer, Pancreatic Cancer, Ovarian Cancer, Colon  and Gastric Cancer Patients show that FOXM1 expression is associated with shorter overall survival (OS) in the majority of most commonly diagnosed cancers. In  gastric cancer, significant association between FOXM1 expression and poor survival is detected in the patients with an advance stage (stage IV)

Figure 2: Kaplan Meier Survival Analysis of FOXM1 mRNA Expression on Overall Survival of Breast Cancer, Lung Cancer, Pancreatic Cancer, Ovarian Cancer, Colon and Gastric Cancer Patients show that FOXM1 expression is associated with shorter overall survival (OS) in the majority of most commonly diagnosed cancers. In gastric cancer, significant association between FOXM1 expression and poor survival is detected in the patients with an advance stage (stage IV)

FOXM1 PROMOTES STEMNESS THROUGH INDUCTION OF KEY TRANSCRIPTION FACTORS

SOX2, a transcription factor and pluripotency marker involved in embryogenesis, has been linked to stemness, cell proliferation, plasticity, and therapy resistance in numerous tumors, including esophageal squamous cancer, gastric cancer, ovarian cancer, medulloblastoma, glioblastoma, lung, and breast cancer [33,40- 47]. Genomic analysis in glioblastoma patients showed that FOXM1 directly binds to the promotor of SOX2, driving its expression. FOXM1 knockdown suppresses SOX2 expression and sensitizes glioblastoma cells to radiation [48]. Studies in in vitro and in vivo tumor models of Triple Negative Breast Cancer (TNBC) demonstrated that FOXM1 plays a role in the regulation of SOX2 via DNMT1/FOXO3a/FOXM1/SOX2 signaling axis and its inhibition suppresses CD44+/CD24- and ALDH+ stem cell population significantly [49]. FOXM1 has also been reported as a regulator of SOX2 in colorectal cancer by inhibiting the promoter activity of SOX2 [50]. Recently, another study revealed the link between the modulatory effect of the FOXM1 on SOX2 axis in colorectal cancer as the inhibition of FOXM1/SOX2 signaling reduced CXCR4, stemness marker expression and spheroid formation [51]. It has also been reported that KMT2A regulates FOXM1 and SOX2 via KLF11, and KMT2A inhibition, leading to decreased spheroid formation ability in human gastric cancer cell lines [52].

NANOG is another transcription factor with its role in pluripotency and differentiation in embryogenesis [53]. NANOG promotes tumor formation, self-renewal, apoptosis, drug resistance, formation of angiogenesis and cancer stemness [54]. A recent study showed that FOXM1 mediated by OTUD7B plays a role in NANOG and SOX2 expression in TNBC cells, and its inhibition suppresses tumor spheroid sizes and expression of stemness markers such as CD44 and EPCAM [55]. In lung cancer, FOXM1 inhibition via FOXM1 shRNA and Genistein combination resulted in suppression of spheroid formation and NANOG expression [56]. Another study revealed FOXM1 and NANOG expressions are regulated by ALKBH5 in cisplatin resistant head and neck squamous cancer [57].

OCT4, a transcription factor that can induce pluripotency, has been shown in various cancers as an oncogenic target and may function in creating complexes with other transcription factors like SOX2, NANOG, and ABCG2 [58]. In the triple negative breast cancer in vitro tumor model, FOXM1 inhibition by shRNA decreased the size and frequency of spheroid formation and suppressed the OCT4 and NANOG expression via the YAP1 signaling pathway [59]. In embryonal carcinoma cells, FOXM1 has been reported to regulate OCT4, and inhibition of FOXM1 directly suppresses OCT4 and NANOG expression levels [60].

FOXM1 Sustains CSC Proliferation and Promotes Invasion and Migration

FOXM1 promotes cancer cell proliferation through its regulatory role in the cell cycle, with dysregulated FOXM1 being associated with increased mitosis [61,62]. Studies by Lou et al. revealed that FOXM1 induces G0/G1 to the S phase switch in Nasopharyngeal Cancer (NPC) cells while promoting in vivo tumor formation, in vitro spheroid formation and expression of stemness-related proteins, ABCG2, SOX2, NANOG and OCT4 [63]. FOXM1 is associated with tumor progression and shorter progression-free patient survival in ovarian cancer; in vitro studies with chemo-resistant IGROV1 ovarian cancer cells showed higher expression of stemness markers, CD44, SOX2 and NANOG, all in correlation with FOXM1 expression. Along with this finding FOXM1 overexpression promoted the same markers, CD44, SOX2, NANOG, and spheroid formation [64]. In the in vivo model of hepatocellular carcinoma (HCC), direct binding of FOXM1 to the promotor region of CD44 has been demonstrated, and FOXM1’s deletion significantly suppressed the tumor nodule formation, specifically the CD44, EPCAM expressing cell population [65]. In a liver cancer model, CD133+/ CD24+ cells were shown to have a higher capacity of stemness and FOXM1 expression. Inhibition of FOXM1 via siRNA in liver cancer stem cells (LCSCs) suppressed ki67 expression and reduced the proliferation, migration and invasion capacity of LCSCs. Also, researchers found that FOXM1 inhibition suppresses protein expression of N-Cadherin, E-Cadherin and Vimentin, which are associated with epithelial-mesenchymal-transition (EMT) [66]. FOXM1 is linked to β-catenin/Wnt pathway regulation and stemness in many studies [67-71]. Su et al. showed that CD133+/CD44+ lung cancer stem cells (LCSCs) have a higher capacity of tumor formation, and FOXM1 is upregulated in that subpopulation. Later, they found that FOXM1 plays a role as a downstream target of the β-catenin/Wnt pathway, and inhibition of FOXM1 via siRNA suppresses migration and invasion ability of LCSCs [71]. CENPU was found highly expressed in cervical cancer patients and in vitro analysis showed that CENPU regulates stemness markers, FOXM1/β-catenin/Wnt signaling, and its inhibition halts migration and invasion, while FOXM1 induction reverses the effects of CENPU inhibition [72]. In colorectal cancer, Valverde et al. used a combination of AEE788, a multiple kinase inhibitor, and Celecoxib, a COX2 inhibitor, and found that it suppresses FOXM1 expression, spheroid formation and migration ability [73].

The complexity of tumorigenesis is not only the result of many intrinsic regulations but also its interaction with the surrounding microenvironment, including the extracellular matrix, immune cells, fibroblasts, and epithelial cells [74]. The role of FOXM1 in CSCs and tumor microenvironment remains to be elucidated. Patient-derived ovarian CSCs (OCSCs) co- cultured in a 3D organotypic system showed higher FOXM1 expression and activity compared to OCSCs cultured without tumor microenvironment components. The FOXM1 inhibitor Thiostrepton reduced the viability of OCSCs and enhanced the effects of the PARP inhibitor Olaparib [75].

FOXM1-MEDIATED DRUG RESISTANCE IN CSCS

Due to their association with reduced therapeutic response, CSCs have been proposed as an essential target to overcome drug resistance [76]. ABC transporters play a role in drug efflux and cause multidrug resistance. Bergamaschi et al. demonstrated that FOXM1 is associated with tamoxifen resistance in ER positive breast cancer by driving the expression of ABC transporter ABCG2 and promoting spheroid formation [77]. In another breast cancer in vitro model using MDA-MB-231 and MCF7 cell lines, GDF-15 expression correlated with FOXM1, SOX2, OCT4, and ABCC5 expressions [78]. In nasopharyngeal cancer (NPC), it has been found that paclitaxel resistance correlates with higher FOXM1, CD44, ALDH1, SOX2, and ABCC5. FOXM1 inhibition with siRNA lowered the expression of ABCC5 and sensitized the NPC to paclitaxel [79]. In high-grade non-serous-epithelial ovarian cancer patients, FOXM1, was found to be highly expressed, and its expression correlated with the tumor grade and inhibition of FOXM1, whereby siRNA sensitized the cells to chemotherapeutic agents such as carboplatin, cisplatin and doxorubicin [80]. In another chemo-resistant ovarian cancer model, researchers showed that chemo resistance is linked to higher stemness capacity, and FOXM1 inhibition reduces stemness while inhibiting resistance to cisplatin [81]. In the pancreatic cancer in vitro model, Trametes Robiniophila Murr (Huaier) which suppresses FOXM1 expression, was shown to inhibit spheroid formation. In an in vivo breast cancer model, FOXM1 inhibition improved the effect of gemcitabine [82]. In the prostate cancer model, Yuan et al. showed that FOXM1 regulates stemness through UHTF1 gene transcription, which has been shown to play a role in docetaxel resistance [83]. Recently, Li et al. showed that another FOX- related protein, FBXO7, regulates stemness via exon Va inclusion of FOXM1. That process promotes a more stabilized version of FOXM, FOXM1c subtype, related to poor patient survival. Notably, inhibition of FBXO7 sensitizes the glioblastoma to temozolomide in vitro and in vivo [84].

FOXM1 INHIBITORS FOR TARGETING CSCS AND CANCER CELLS

Given its regulatory role on tumorigenesis, resistance to conventional therapies and influence on cancer stemness, FOXM1 represents a promising target for cancer treatment. Currently, no FDA-approved therapies specifically target FOXM1. Although several FOXM1 inhibitors have been identified, none have progressed to clinical trials due to a lack of potency and selectivity. Therefore, there is an urgent need for the development of potent, highly selective, and safe FOXM1 inhibitors. Bu et al. identified M1-20, an interfering protein and a FOXM1 transcription inhibitor, demonstrating its inhibitory effects on cervical cancer, TNBC, ER+ breast cancer, sarcoma and lung cancer cell lines. They found that FOXM1 inhibition suppresses the expression of stem cell markers ALDH1 and CD44 expression and enhances chemotherapy efficacy [85]. In osteosarcoma cells, small molecule inhibitors NSM00158 and NSC95397 showed regulatory effects on in vitro and in vivo tumor growth via CtBP1/FOXM1/MDR1 signaling axis, sensitizing the tumors to cisplatin chemotherapy [86]. Roca et al., demonstrated that Domatinostat (4SC-202), a selective HDAC small molecule inhibitor in clinical trials, suppresses FOXM1 mRNA expression and spheroid formation while sensitizing pancreatic cancer models to chemotherapy [87].

It was also found that AURKA, Aurora Kinase A, and FOXM1 overexpression promote stemness in breast cancer through a positive feedback mechanism. The novel AURKA inhibitor AKI603, in combination with the FOXM1 inhibitor Thiostrepton, synergistically suppressed in vitro spheroid formation and in vivo tumor formation, suggesting that combining FOXM1 inhibitors with AURORA inhibitors may overcome therapy resistance [88]. In a liver cancer in vitro model, Thiostrepton reduced sphere formation and the expression of CD44, SOX2 in regorafenib-resistant cells, indicating its potential to target stemness signatures [89]. Joshi et al., showed that Siomycin-A, a FOXM1 inhibitor, significantly reduced the spheroid formation capacity of cells derived from in vivo glioblastoma tumors compared to control groups. Siomycin-A combined with the chemotherapy agent Temozolomide suppressed in vivo tumor growth and reduced drug resistance in glioblastoma stem cell- induced tumors [90]. In hepatocellular carcinoma (HCC) in vitro models, Alfa-fetoprotein (AFP)-positive cells showed high expression of FOXM1. Carfilzomib, a proteasome inhibitor, inhibited the expression of FOXM1 and AFP. In the in vivo model, Carfilzomib combination with DC10, an anti-mouse VEGFR2 antibody, reduced the viability of tumors derived from AFP- positive cells [91]. In addition, Zhang et al. showed the effect of M-138, a recombinant protein constructed explicitly for FOMX1 N terminus inhibitory domain activation, on BC, HCC, lung cancer, and cervical adenocarcinoma cell lines. M-138 disturbed the interaction between FOXM1 and SMAD3, suppressed the in vivo tumor growth and OCT4, ALDH1 expression [92] [Table 1].

Table 1: FOXM1 Inhibitors with their effects on Cancer Stemness

Inhibitors-Drugs

Signaling Axis

Regulatory Effect

Cancer Type

Ref.

5-AZAC

FOXO3a/FOXM1/SOX2

Suppression Of Spheroid Formation and Migration

TNBC

[73]

AEE788 and Celecoxib

FOXM1 / -catenin

Suppression of CD44+/CD24- and ALDH+ cells

Colorectal Cancer

[73]

Carfilzomib

-

Suppression of AFP+ cells

HCC

[91]

Domatinostat

-

Suppression of Spheroid Formation, Reduced Chemo- resistance

Pancreatic Cancer

[87]

M1-20

-

Suppression of ALDH1 and CD44 expressions

CC, TNBC, ER+ breast Cancer, Sarcoma, Lung Cancer

[85]

M-138

FOXM1/SMAD3

Inhibition of OCT4, ALDH1

BC, HCC, Lung Cancer, Cervical Adeno-carcinoma

[92]

NSM00158 / NSC95397

CtBP1/FOXM1/MDR1

Reduced Tumor Growth and Chemo-resistance

Osteosarcoma

[86]

Siomycin-A

FOXM1/MELK

Suppression of Spheroid Formation, Reduced Chemo- resistance

Glioblastoma

[90]

Thiostrepton

FOXM1-DNA-binding Domain Inhibitor

Suppression of Spheroid Formation, In-vivo Tumor Formation and CD44, and SOX2 Expression

Breast Cancer, Liver Cancer

[88,89,93]

Trametes Robiniophila Murr (huaier)

Nuclear Translocation of FOXM1

Suppression of Spheroid Formation, Reduced Chemo- resistance

Pancreatic Cancer

[82]

CONCLUDING REMARKS AND FUTURE PROSPECTS

FOXM1 is a commonly overexpressed oncogenic transcription factor linked to poor patient survival and is an emerging molecular target for cancer treatment, including CSCs. Many preclinical studies underline FOXM1’s regulatory effect on key transcription factors related to pluripotency, stemness, tumorigenesis, migration, invasion, epithelial mesenchymal transition, and drug resistance. Most notably, FOXM1 inhibition enhances the effect of chemotherapy and radiotherapy across various cancer models,making it a critical player in overcoming therapy resistance and suppressing tumor growth and progression. However, its effects on cancer stemness and the tumor microenvironment remains to be fully elucidated. The use of 3D-organoid models could provide clearer insights into its role in the near future. Additionally, the role of FOXM1 subtypes in stemness capacity is still unclear. Despite its potential as a molecular target in many cancer types, no FDA-approved targeted therapy for FOXM1 exists. Given its oncogenic importance, there is an urgent need to develop effective FOXM1 inhibitors. The identification of safe and effective FOXM1 inhibitors and their translation to the clinic may significantly impact the treatment of FOXM1-driven cancers and improve patient survival.

REFERENCES
  1. Kochanek KD, Murphy SL, Xu J, Arias E. Mortality in the United States, 2022 Key findings Data from the National Vital Statistics System. 2022.
  2. Siegel RL, Giaquinto AN, Ahmedin J. Cancer statistics, 2024. CA Cancer J Clin. 2024; 74: 12-49.
  3. Debela DT, Muzazu SGY, Heraro KD, Ndalama MT, Mesele BW, Haile DC, et al. New approaches and procedures for cancer treatment: Current perspectives. SAGE Open Med. 2021; 9.
  4. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation.Cell. 2011; 144: 646-674.
  5. Vasan N, Baselga J, Hyman DM. A view on drug resistance in cancer. Nature. 2019; 575: 299-309.
  6. Wu S, Zhu W, Thompson P, Hannun YA. Evaluating intrinsic and non-intrinsic cancer risk factors. Nature Commun. 2018; 9: 3490.
  7. Zhang S, Xiao X, Yi Y, Wang X, Zhu L, Shen Y, et al. Tumor initiation and early tumorigenesis: molecular mechanisms and interventional targets. Signal Transduct Targ Ther. 2024; 9: 1-36.
  8. Loh JJ, Ma S. Hallmarks of cancer stemness. Cell Stem Cell. 2024; 31: 617-639.
  9. Fumagalli A, Oost KC, Kester L, Morgner J, Bornes L, Bruens L, et al. Plasticity of Lgr5-Negative Cancer Cells Drives Metastasis in Colorectal Cancer. Cell Stem Cell. 2020; 26: 569-578.
  10. Leng Z, Xia Q, Chen J, Li Y, Xu J, Zhao E, et al. Lgr5+CD44+EpCAM+ Strictly Defines Cancer Stem Cells in Human Colorectal Cancer. Cell Physiol Biochem. 2018; 46: 860-872.
  11. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003; 100: 3983-3988.
  12. Lapidot T, Sirard C, Vormoor J. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994; 367: 645-648.
  13. Bonnet D, Dick J. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997; 3: 730-737.
  14. Li W, Ma H, Zhang J, Zhu L, Wang C, Yang Y. Unraveling the roles of CD44/CD24 and ALDH1 as cancer stem cell markers in tumorigenesis and metastasis. Sci Rep. 2018; 8: 4276.
  15. Luo Y, Dallaglio K, Chen Y, Robinson WA, Robinson SE, McCarter MD, et al. ALDH1A isozymes are markers of human melanoma stem cells and potential therapeutic targets. Stem Cells. 2012; 30: 2100-2113.
  16. Markowska A, Kojs Z, Twardawa D, Pietras J, Markowska J. Selected markers of ovarian cancer and their relation to targeted therapy (Review). Exp Ther Med. 2024; 27: 236.
  17. Monzani E, Facchetti F, Galmozzi E, Corsini E, Benetti A, Cavazzin C, et al. Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential. Eur J Cancer. 2007; 43: 935-946.
  18. Pospieszna J, Dams-Kozlowska H, Udomsak W, Murias M, Kucinska M. Unmasking the Deceptive Nature of Cancer Stem Cells: The Role of CD133 in Revealing Their Secrets. Int J Mol Sci. 2023; 24: 10910.
  19. Jeter CR, Yang T, Wang J, Chao HP, Tang DG. NANOG in cancer stem cells and tumor development: An update and outstanding questions. 2015; 33: 2381-2390.
  20. Zhao W, Li Y, Zhang X. Stemness-Related Markers in Cancer. Cancer Transl Med. 2017; 3: 87-95.
  21. Auffinger B, Tobias AL, Han Y, Lee G, Guo D, Dey M, et al. Conversion of differentiated cancer cells into cancer stem-like cells in a glioblastoma model after primary chemotherapy. Cell Death Differ. 2014; 21: 1119-1131.
  22. Makena MR, Ranjan A, Thirumala V, Reddy AP. Cancer stem cells: Road to therapeutic resistance and strategies to overcome resistance. Biochim Biophys Acta Mol Basis Dis. 2020; 1866: 165339.
  23. Lagadec C, Vlashi E, Della Donna L, Dekmezian C, Pajonk F. Radiation- induced reprogramming of breast cancer cells. Stem Cells. 2012; 30: 833-844
  24. Kaestner KH, Knö W, Martínez DE. Unified nomenclature for the winged helix/forkhead transcription factors. Genes Dev. 2000; 14: 142-146.
  25. Jackson BC, Carpenter C, Nebert DW, Vasiliou V. Update of human and mouse forkhead box (FOX) gene families. Hum Genomics. 2010; 4: 345-352.
  26. Zhu J, Wei Y, Deng F, Zhou Y, Yang Z, Ma Y. The Role of FOXA1 in Human Normal Development and Its Functions in Sex Hormone- Related Cancers. Front Biosci Landmark (Ed). 2024; 29: 225.
  27. Xu J, Wang K, Zhang Z, Xue D, Li W, Pan Z. The Role of Forkhead Box Family in Bone Metabolism and Diseases. Front Pharmacol. 2021; 12: 772237.
  28. Cheng M, Nie Y, Song M, Chen F, Yu Y. Forkhead box O proteins: steering the course of stem cell fate. Cell Regen. 2024; 13: 7.
  29. Ahn JI, Lee KH, Shin DM, Shim JW, Kim CM, Kim H, et al. Temporal expression changes during differentiation of neural stem cells derived from mouse embryonic stem cell. J Cell Biochem. 2004; 93: 563-578.
  30. Krupczak-Hollis K, Wang X, Kalinichenko VV, Gusarova GA, Wang IC, Dennewitz MB, et al. The mouse Forkhead Box m1 transcription factor is essential for hepatoblast mitosis and development of intrahepatic bile ducts and vessels during liver morphogenesis. Dev Biol. 2004; 276: 74-88.
  31. Peñailillo R, Velásquez V, Acuña-Gallardo S, García F, Sánchez M, Nardocci G, et al. FOXM1 Participates in Trophoblast Migration and Early Trophoblast Invasion: Potential Role in Blastocyst Implantation. Int J Mol Sci. 2024; 25: 1678.
  32. Kim IM, Ramakrishna S, Gusarova GA, Yoder HM, Costa RH, Kalinichenko V V. The forkhead box m1 transcription factor is essential for embryonic development of pulmonary vasculature. J Biol Chem. 2005; 280: 22278-22286.
  33. Hamurcu Z, Ashour A, Kahraman N, Ozpolat B. FOXM1 regulates expression of eukaryotic elongation factor 2 kinase and promotes proliferation, invasion and tumorgenesis of human triple negative breast cancer cells. Oncotarget. 2016; 7: 16619-16635.
  34. Chen L, Wu M, Ji C, Yuan M, Liu C, Yin Q. Silencing transcription factor FOXM1 represses proliferation, migration, and invasion while inducing apoptosis of liver cancer stem cells by regulating the expression of ALDH2. IUBMB Life. 2020; 72: 285-95.
  35. Li L, Wu D, Yu Q, Li L, Wu P. Prognostic value of FOXM1 in solid tumors: a systematic review and meta-analysis. Oncotarget. 2017; 8: 32298-32308.
  36. Pandit B, Halasi M, Gartel AL. Cell Cycle p53 negatively regulates expression of FoxM1. Cell Cycle. 2009; 8: 3425-3427.
  37. Chen X, Zhang T, Su W, Dou Z, Zhao D, Jin X, et al. Mutant p53 in cancer: from molecular mechanism to therapeutic modulation. Cell Death Dis. 2022; 13: 974.
  38. Li L, Wu D, Yu Q, Li L, Wu P. Prognostic value of FOXM1 in solid tumors: a systematic review and meta-analysis. Oncotarget. 2017; 8: 32298-32308.
  39. Wei G, Yang X, Lu H, Zhang L, Wei Y, Li H, et al. Prognostic value and immunological role of FOXM1 in human solid tumors. Aging (Albany NY). 2022; 14: 9128-9148.
  40. Gao H, Teng C, Huang W, Peng J, Wang C. SOX2 Promotes the Epithelial to Mesenchymal Transition of Esophageal Squamous Cells by Modulating Slug Expression through the Activation of STAT3/ HIF-α Signaling. Int J Mol Sci. 2015; 16: 21643-21657.
  41. Chen Y, Huang Y, Zhu L, Chen M, Huang Y, Zhang J, et al. SOX2 inhibits metastasis in gastric cancer. J Cancer Res Clin Oncol. 2016; 142: 1221-1230.
  42. Bareiss PM, Paczulla A, Wang H, Schairer R, Wiehr S, Kohlhofer U, et al. SOX2 expression associates with stem cell state in human ovarian carcinoma. Cancer Res. 2013; 73: 5544-5555.
  43. Cox JL, Wilder PJ, Desler M, Rizzino A. Elevating SOX2 levels deleteriously affects the growth of medulloblastoma and glioblastoma cells. PLoS One. 2012; 7: e44087.
  44. Nakatsugawa M, Takahashi A, Hirohashi Y, Torigoe T, Inoda S, Murase M, et al. SOX2 is overexpressed in stem-like cells of human lung adenocarcinoma and augments the tumorigenicity. Lab Invest. 2011; 91: 1796-1804.
  45. Zhou HY, Katsman Y, Dhaliwal NK, Davidson S, Macpherson NN, Sakthidevi M, et al. A Sox2 distal enhancer cluster regulates embryonic stem cell differentiation potential. Genes Dev. 2014; 28: 2699-2711.
  46. Jeon HM, Sohn YW, Oh SY, Kim SH, Beck S, Kim S, et al. ID4 imparts chemoresistance and cancer stemness to glioma cells by derepressing miR-9*-mediated suppression of SOX2. Cancer Res. 2011; 71: 3410-3421.
  47. Alonso MM, Diez-Valle R, Manterola L, Rubio A, Liu D, Cortes-Santiago N, et al. Genetic and Epigenetic Modifications of Sox2 Contribute to the Invasive Phenotype of Malignant Gliomas. PLoS One. 2011; 6: 26740.
  48. Lee Y, Kim KH, Kim DG, Cho HJ, Kim Y, Rheey J, et al. FoxM1 Promotes Stemness and Radio-Resistance of Glioblastoma by Regulating the Master Stem Cell Regulator Sox2. PLoS One. 2015; 10: e0137703.
  49. Liu H, Song Y, Qiu H, Liu Y, Luo K, Yi Y, et al. Downregulation of FOXO3a by DNMT1 promotes breast cancer stem cell properties and tumorigenesis. Cell Death Differ. 2020; 27: 966-983.
  50. Fu Y, Bai C, Wang S, Chen D, Zhang P, Wei H, et al. AKT1 phosphorylates RBM17 to promote Sox2 transcription by modulating alternative splicing of FOXM1 to enhance cancer stem cell properties in colorectal cancer cells. FASEB J. 2023; 37: e22707.
  51. Peng H, Ye T, Deng L, Yang X, Li Q, et al. Activin and Hepatocyte Growth Factor Promotes Colorectal Cancer Stemness and Metastasis through FOXM1/SOX2/CXCR4 Signaling. Gut Liver. 2024; 18: 476.
  52. Deng C, Ye C, Liao X, Zhou F, Shi Y, et al. KMT2A maintains stemness of gastric cancer cells through regulating Wnt/β-catenin signaling- activated transcriptional factor KLF11. Open Med. 2023; 18: 20230764.
  53. Silva J, Nichols J, Theunissen TW, Guo G, van Oosten AL, et al. Nanog Is the Gateway to the Pluripotent Ground State. Cell. 2009; 138: 722- 737.
  54. Najafzadeh B, Asadzadeh Z, Motafakker Azad R, Mokhtarzadeh A, Baghbanzadeh A, et al. The oncogenic potential of NANOG: An important cancer induction mediator. J Cell Physiol. 2021; 236: 2443- 2458.
  55. Wang H, Han S, Xiao J, Fu X, Chen W, et al. OTUD7B knockdown inhibits the proliferation and stemness of breast cancer cells by destabilizing FOXM1. Oncol Lett. 2024; 27: 102.
  56. Fu Z, Cao X, Liu L, Cao X, Cui Y, et al. Genistein inhibits lung cancer cell stem like characteristics by modulating MnSOD and FoxM1 expression. Oncol Lett. 2020; 20: 2506-2515.
  57. Shriwas O, Priyadarshini M, Samal SK, Rath R, Panda S, et al. DDX3 modulates cisplatin resistance in OSCC through ALKBH5-mediated m6A-demethylation of FOXM1 and NANOG. Apoptosis. 2020; 25: 233-246.
  58. Zhang Q, Han Z, Zhu Y, Chen J, Li W. The Role and Specific Mechanism of OCT4 in Cancer Stem Cells: A Review. Int J Stem Cells. 2020; 13: 312-325.
  59. Sun HL, Men JR, Liu HY, Liu MY, Zhang HS. FOXM1 facilitates breast cancer cell stemness and migration in YAP1-dependent manner. Arch Biochem Biophys. 2020; 685: 108349.
  60. Xie Z, Tan G, Ding M, Dong D, Chen T, et al. Foxm1 transcription factor is required for maintenance of pluripotency of P19 embryonal carcinoma cells. Nucleic Acids Res. 2010; 38: 8027-8038.
  61. Chen X, Müller GA, Quaas M, Fischer M, Han N, et al. The forkhead transcription factor FOXM1 controls cell cycle-dependent gene expression through an atypical chromatin binding mechanism. Mol Cell Biol. 2013; 33: 227-236.
  62. Kalathil D, John S, Nair AS. FOXM1 and Cancer: Faulty Cellular Signaling Derails Homeostasis. Front Oncol. 2020; 10: 626836.
  63. Luo W, Gao F, Li S, Liu L. FOXM1 promotes cell proliferation, invasion, and stem cell properties in nasopharyngeal carcinoma. Front Oncol. 2018; 8: 483.
  64. Chiu WT, Huang YF, Tsai HY, Chen CC, Chang CH, et al. FOXM1 confers to epithelial-mesenchymal transition, stemness and chemoresistance in epithelial ovarian carcinoma cells. Oncotarget. 2014; 6: 2349-2365.
  65. Kopanja D, Pandey A, Kiefer M, Wang Z, Chandan N, et al. Essential roles of FoxM1 in Ras-induced liver cancer progression and in cancer cells with stem cell features. J Hepatol. 2015; 63: 429-436.
  66. Chen L, Wu M, Ji C, Yuan M, Liu C, et al. Silencing transcription factor FOXM1 represses proliferation, migration, and invasion while inducing apoptosis of liver cancer stem cells by regulating the expression of ALDH2. IUBMB Life. 2020; 72: 285-295.
  67. Gong A, Huang S. FoxM1 and Wnt/β-catenin signaling in glioma stem cells. Cancer Res. 2012; 72: 5658-5662.
  68. Bowman A, Nusse R. Location, Location, Location: FoxM1 Mediates β-Catenin Nuclear Translocation and Promotes Glioma Tumorigenesis. Cancer Cell. 2011; 20: 415-416.
  69. Zhang N, Wei P, Gong A, Chiu WT, Lee H Te, et al. FoxM1 Promotes β-Catenin Nuclear Localization and Controls Wnt Target-Gene Expression and Glioma Tumorigenesis. Cancer Cell. 2011; 20: 427-442.
  70. Xie H, Miao N, Xu D, Zhou Z, Ni J, et al. FoxM1 promotes Wnt/β-catenin pathway activation and renal fibrosis via transcriptionally regulating multi-Wnts expressions. J Cell Mol Med. 2021; 25: 1958-1971.
  71. Su J, Wu S, Wu H, Li L, Guo T. CD44 is functionally crucial for driving lung cancer stem cells metastasis through Wnt/β-catenin-FoxM1- Twist signaling. Mol Carcinog. 2016; 55: 1962-1973.
  72. Zhang X, Wang M, Zhang Y, Yang J, Duan W. Knockdown of CENPU inhibits cervical cancer cell migration and stemness through the FOXM1/Wnt/β-catenin pathway. Tissue Cell. 2023; 81: 102009.
  73. Valverde A, Peñarando J, Cañas A, López-Sánchez LM, Conde F, et al. Simultaneous Inhibition of EGFR/VEGFR and Cyclooxygenase-2 Targets Stemness-Related Pathways in Colorectal Cancer Cells. PLoS One. 2015; 10: e0131363.
  74. Anderson NM, Simon MC. The tumor microenvironment. Curr Biol. 2020; 30: R921-R925.
  75. Battistini C, Kenny HA, Zambuto M, Nieddu V, Melocchi V, et al. Tumor microenvironment-induced FOXM1 regulates ovarian cancer stemness. Cell Death & Disease. 2024; 15: 370.
  76. Cho Y, Kim YK. Cancer Stem Cells as a Potential Target to Overcome Multidrug Resistance. Front Oncol. 2020; 10: 764.
  77. Bergamaschi A, Madak-Erdogan Z, Kim YJ, Choi Y La, Lu H, et al. The forkhead transcription factor FOXM1 promotes endocrine resistance and invasiveness in estrogen receptor-positive breast cancer by expansion of stem-like cancer cells. Breast Cancer Res. 2014; 16: 1-18.
  78. Modi A, Purohit P, Roy D, Vishnoi JR, Pareek P, et al. FOXM1 mediates GDF-15 dependent stemness and intrinsic drug resistance in breast cancer. Mol Biol Rep. 2022; 49: 2877-2888.
  79. Hou Y, Zhu Q, Li Z, Peng Y, Yu X, et al. The FOXM1–ABCC5 axis contributes to paclitaxel resistance in nasopharyngeal carcinoma cells. Cell Death Dis. 2017; 8: e2659.
  80. Tassi RA, Todeschini P, Siegel ER, Calza S, Cappella P, et al. FOXM1 expression is significantly associated with chemotherapy resistance and adverse prognosis in non-serous epithelial ovarian cancer patients. J Exp Clin Cancer Res. 2017; 36: 63.
  81. Chiu W-T, Huang Y-F, Tsai H-Y, Chen C-C, Chang C-H, Huang S-C, et al. FOXM1 confers to epithelial-mesenchymal transition, stemness and chemoresistance in epithelial ovarian carcinoma cells. Oncotarget. 2014; 6: 2349-2365.
  82. Wang Q, Gong M, Liu R, Mo J, Bai R, An R, et al. Huaier enhances the tumor-killing effect and reverses gemcitabine-induced stemness by suppressing FoxM1. Phytomed. 2024; 129: 155656.
  83. Yuan B, Liu Y, Yu X, Yin L, Peng Y, Gao Y, et al. FOXM1 contributes to taxane resistance by regulating UHRF1-controlled cancer cell stemness. Cell Death Dis. 2018; 9: 1-11.
  84. Li S, Chen Y, Xie Y, Zhan H, Zeng Y, Zeng K, et al. FBXO7 Confers Mesenchymal Properties and Chemoresistance in Glioblastoma by Controlling Rbfox2-Mediated Alternative Splicing. Adv Sci (Weinh). 2023; 10: e2303561.
  85. Bu H, Lan X, Cheng H, Pei C, Ouyang M, Chen Y, et al. Development of an interfering peptide M1-20 with potent anti-cancer effects by targeting FOXM1. Cell Death Dis. 2023; 14: 1-10.
  86. Chen X, Zhang Q, Dang X, Song T, Wang Y, Yu Z, et al. Targeting the CtBP1-FOXM1 transcriptional complex with small molecules to overcome MDR1-mediated chemoresistance in osteosarcoma cancer stem cells. J Cancer. 2021; 12: 482-497.
  87. Roca MS, Moccia T, Iannelli F, Testa C, Vitagliano C, Minopoli M, et al. HDAC class I inhibitor domatinostat sensitizes pancreatic cancer to chemotherapy by targeting cancer stem cell compartment via FOXM1 modulation. J Exp Clin Cancer Res. 2022; 41: 83.
  88. Yang N, Wang C, Wang Z, Zona S, Lin SX, Wang X, et al. FOXM1 recruits nuclear Aurora kinase A to participate in a positive feedback loop essential for the self-renewal of breast cancer stem cells. Oncogene. 2017; 36: 3428-3440.
  89. Wuputra K, Hsiao PJ, Chang WT, Wu PH, Chen LA, Huang JW, et al. FOXM1-CD44 Signaling Is Critical for the Acquisition of Regorafenib Resistance in Human Liver Cancer Cells. Int J Mol Sci. 2022; 23: 7782.
  90. Joshi K, Banasavadi-Siddegowda Y, Mo X, Kim SH, Mao P, Kig C, et al. MELK-Dependent FOXM1 Phosphorylation is Essential for Proliferation of Glioma Stem Cells. Stem Cells. 2013; 31: 1051-1063.
  91. Li R, Okada H, Yamashita T, Nio K, Chen H, Li Y, et al. FOXM1 Is a Novel Molecular Target of AFP-Positive Hepatocellular Carcinoma Abrogated by Proteasome Inhibition. Int J Mol Sci. 2022; 23: 8305.
  92. Zhang Z, Bu H, Yu J, Chen Y, Pei C, Yu L, et al. The cell-penetrating FOXM1 N-terminus (M1-138) demonstrates potent inhibitory effects on cancer cells by targeting FOXM1 and FOXM1-interacting factor SMAD3. Theranostics. 2019; 9: 2882-3896.
  93. Kwok JMM, Myatt SS, Marson CM, Coombes RC, Constantinidou D, Lam EWF. Thiostrepton selectively targets breast cancer cells through inhibition of forkhead box M1 expression. Mol Cancer Ther. 2008; 7: 2022-2032.

Biltekin E, Ozpolat B (2024) FOXM1: A Promising Target for Cancer Stemness. J Cancer Biol Res 11(2): 1146.

Received : 03 Jul 2024
Accepted : 31 Jul 2024
Published : 31 Jul 2024
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