Loading

Annals of Food Processing and Preservation

microRNAs as Therapeutic Targets in Diagnosis, Prognosis, and Treatment of Pancreatic Cancer: A Mini Review

Mini Review | Open Access | Volume 7 | Issue 1

  • 1. Department of Biology, Science and Research Branch, Islamic Azad University, Iran
  • 2. Department of Biological Sciences, University of Texas at Dallas, USA
  • 3. School of Nutrition and Food Science, Isfahan University of Medical Sciences, Iran
  • 4. Department of Biological Sciences, University of North Texas, USA
  • 5. Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Iran
+ Show More - Show Less
Corresponding Authors
Marjan Ganjali Dashti, Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA, Tel: 693-638-905
Abstract

MicroRNAs (miRNA) are potent targets for cancer therapy. Altered miRNA expression is commonly associated with cancer stem cells and EMT (epithelial-to-mesenchymal transition) phenotypes which are hallmarks of tumor initiation, progression, and metastasis. These non-coding RNAs bind to the 3′ UTR of target mRNAs and degrade them or inhibit their translation; they are potent diagnostic, prognostic, and therapeutic molecules. Although some miRNAs can prevent tumor growth, some others can promote cancer. For cancer treatment, tumor suppressor miRNAs should be up-regulated whereas oncomirs should be down-regulated. Furthermore, studies show that nature-derived components such as oridonin, curcumin, isoflavone, I3C, DIM, EGCG, and resveratrol can target several miRNAs simultaneously and inhibit cancer growth, induce apoptosis, reverse EMT to MET phenotypes, and eliminate CSCs or EMT phenotypic cells (which cause tumor recurrence and resistance to different conventional therapies). This mini review will discuss the list of nature-derived components which improve the overall survival of cancer patients.

Keywords

microRNAs,  Pancreatic cancer,  Treatment,  Nature-derived components

INTRODUCTION

Due to its poor prognosis, inadequate detection, and treatment, pancreatic cancer is one of the tumor types with a high fatality rate [1]. There are numerous therapeutic strategies against this cancer, with chemotherapy being the most popular treatment. However, chemotherapy is ineffective against pancreatic cancer because of the unique tumor microenvironment, which leads to reduced drug efficacy. Another popular therapeutic method is radiotherapy, which has benefits for local tumors but does not work for incurable pancreatic malignancies [2]. Surgery is another option for some malignancies, but because 80–85% of patients are ineligible, it does not achieve the desired outcome for this malignancy [3,4]. Precise knowledge about some of the crucial intracellular processes in cancer cells can help us to find more effective treatments.

One of the most significant intracellular agents is the microRNA family. Endogenous non-coding single-strand RNAs with an average length of 19 to 23 ribonucleotides are referred to as microRNAs (miRNAs) [5]. Their mode of action involves complementarity aligning with the target messenger RNA (mRNA) sequences to reduce their RNA expression levels, which in turn prevents them from being translated or causes the mRNAs to be degraded. Most coding genes are regulated by miRNAs. As a result, they are useful targets for researchers and can aid medical professionals in prognosis, diagnosis, and therapy.

CHARACTERISTICS OF MIRNAS

One of the mystifying characteristics of miRNA is that a single miRNA can target many mRNAs and the same mRNA can be the target of multiple miRNAs. For instance, miR-145 controls a variety of mRNAs, including mucin 13, the TGF-â receptor, SMAD2, and NEDD9 (neural precursor cell expressed developmentally downregulated 9), to reduce the growth and metastasis of pancreatic cancer and to increase the chemosensitivity to gemcitabine (a chemotherapy drug) [6-8]. A candidate target, slug mRNA, may also be suppressed by miR-34 and miR-203a-3p [9]. So, miRNAs contribute to a puzzling and huge network with mRNAs and their targeting is not straightforward. This limits their clinical application. miRNAs are implicated in chemotherapy and drug resistance and understanding their involvement can help clinicians and patients experience better therapeutic outcomes [10].

TARGETING MIRNAS IN PANCREATIC CANCER AS A THERAPEUTIC OPTION

to two main groups: tumor suppressors and oncogenic miRNAs oncomirs. The role of miRNAs tightly depends on the function of their target sequences. When the natural balance between these two fundamental groups breaks, the onset of different conditions occurs [11]. It is postulated that several anticancer actions, such as proliferation, invasion inhibition and increased apoptosis and chemosensitivity can be generated in cancer cells by decreasing the oncomir levels and activating tumor suppressor miRNAs.

TUMOR SUPPRESSOR MIRNAS IN PANCREATIC CANCER

The tumor suppressor miRNA group can degrade target mRNAs to prevent the progression of pancreatic cancer. Therefore, a medical application of these miRNAs is to overexpress them. These miRNAs are associated with upregulating apoptosis, controlling cell cycle mediators, inhibiting oncogenic transcription factors, inhibiting angiogenesis, and increasing chemosensitivity. For instance, miR-455-3p suppresses pancreatic cancer progression by blocking the Wnt/ catenin signaling pathway and promoting these cells’ death by up-regulating the expression of Bcl-2 and Bax proteins, which play a key role in the activation of caspases and apoptosis [5,12]. Other tumor suppressors like miR-373-3p and miR708 increase chemosensitivity. miR-373-3p decreases Cyclin D2 expression, enhances gemcitabine sensitivity (a chemotherapeutic agent), and prevents the growth of gemcitabine-resistant pancreatic cancer cells [13]. miR708 works as a proliferation and chemoresistance inhibitor that suppresses surviving expression [14]. miRNA-145, prevents cancer growth and angiogenesis by angiopoietin-2 (Ang-2) down-regulation while Let-7 prevents the progression of this cancer by up-regulation of cytokine signaling 3 (SOCS3) suppressors to block the phosphorylation of STAT3.

Tumor suppressor miRNAs can also work by controlling other signaling pathways. For instance, miR-34a promotes apoptosis and inhibits metastasis by blocking the expression of Snail1 and Notch 1. However, miR-34a is up-regulated when Notch 1 is down-regulated, creating a feedback loop between the two [15]. Additionally, miR-34 can improve the chemosensitivity of pancreatic tumor cells to gemcitabine gemcitabine as a chemotherapy drug against different types of cancer and induce apoptosis by down-regulating Slug [9,16]. It contributes to pancreatic CSCs’ capacity for self-renewal despite their resistance to a variety of conventional treatments [17]. miR-203a-3p also suppresses Slug and inhibits the proliferation of pancreatic cancer cells and the epithelial to mesenchymal transition (EMT) [9]. EMT is crucial in cancer metastasis. In this regard, focusing on the up-regulation of miRNA candidates involved with suppressing EMT is crucial to stopping the metastasis of cancer. Another tumor suppressor miRNA is MiR-16-1, located on the 13q14.3 locus, and its deletion is reported in B-cells in 65% of chronic lymphocytic leukemia patients [18]. Any decrease in miR-15/16 leads to deficiency of DROSHA processor and elevated activity of histone deacetylases. It leads to the epigenetic silencing of several tumor suppressor genes. Expression of miR16-1 blocks cell proliferation and promotes apoptosis [19]. In NB4 cells transduced with lentiviral miR-16-1, the proliferation and general activity of NB4 cells decreased significantly. But the expression of two important apoptosis-associated genes, Bax and Bcl-2 was like non-treated NB4 cells. Also, miR-16-1 arrests treated NB4 cells in the G1 phase and elevate caspase-3 activity significantly, which can lead to apoptosis. Based on this data, miR-16-1 has medical effects on leukemia [19]. Another study showed that miRNA-20a is overexpressed in various malignancies, including pancreatic, lung, and B-cell lymphomas, and it negatively regulates the E2F TF1 (E2F1), Stat3, and cyclin D1 genes, it can also be regarded as an oncogene [17,20].

ONCOGENIC MIRNAS

Oncogenic miRNAs act in the reverse direction of tumor suppressor miRNAs and promote the progression of cancer, metastasis, and drug-resistance. For example, the miR-21 gene is over-expressed in a variety of human malignancies like pancreatic, lung, liver, and esophageal cancers. It promotes tumor growth, invasion, and migration through targeting some tumor suppressors such as PDCD4, and PTEN (tumor-suppressing phosphatase and tensin homolog), which regulate intracellular signaling pathways, and modulate the cell microenvironment [21,22]. Over-expressed miR-21 in cancer-associated fibroblasts (CAF) can promote metastasis and gemcitabine chemo-resistance by enhancing, level of platelet-derived growth factor (PDGF) as well as matrix metalloproteinases such as MMP-3 and MMP-9 [22]. A significant level of miR-21 also increases 5-fluorouracil (5-FU, a chemotherapeutic agent) resistance [23].

Additionally, in pancreatic cancers, a positive feedback loop has been seen between miR-21 and the epidermal growth factor (EGF) pathway [24]. EGF causes miR-21 to be overexpressed, whereas miR-21 enhances EGF function by inhibiting EGF inhibitors. The activation of additional intracellular cascades, such as the MAPK/ERK and PI3K/AKT signaling pathways, as well as the suppression of apoptosis via Sprouty2 inhibition, are additional ways that miR-21 can promote cell proliferation. One of miR-21’s roles is to reverse the relationship with the Von HippelLindau tumor suppressor [25]. Like a lot of molecules, miR-21 plays two different intracellular roles as it can work as a tumor suppressor miRNA too. For example, it inhibits pancreatic cancer cell proliferation by blocking the HIF1a/VEGF pathway and the expression of some kinds of matrix metalloproteinases such as MMP-2 and MMP-9[12]. In general, overexpression of miR21 is associated with worse remission rates, progression-free survival, more metastatic lymph-nodes and less differentiated tumors [17,26]. Based on some research, it can be considered an appropriate prognostic indicator for chemotherapy classification and a target in the treatment of pancreatic cancer [26]. miR155 is another oncomir that is introduced as a major molecule in pancreatic cancer progression by Foxo3a down-regulation which leads to Reactive Oxygen Species (ROS) induction [27]. Furthermore, miR-155 decreases the level of cytokine signaling 1 and SOCS3 suppressing molecules which leads to STAT3 activation and subsequently induces pancreatic cancer cells proliferation and metastasis [28,29]. Based on these studies, as its role is solely oncogenic, it is promising as a medical target.

Based on some studies, the miR-200 family plays a key role in tumor initiation, metastasis, and the prognosis of pancreatic cancer [30]. In EMT, the expression of the mir-200 family is down-regulated or absent. After pancreatic duct cell carcinoma surgery, the EMT is linked to a lower survival rate. Additionally, mir-200 and let-7 are down-regulated in pancreatic cancer patients, which is related to their resistance to gemcitabine. ZEB1 and vimentin, which are EMT inducers, are down-regulated by the mir-200 family whereas cadherin 1 is up-regulated. Targeting EMTs or CSCs with miRNA can be a medicinal strategy because they promote tumor recurrence and metastasis [17]. miR-320a significantly is up-regulated in pancreatic cancer and leads to the proliferation, intravasation, extravasation, metastasis, and EMT. It binds to the 3′UTR of programmed cell death protein 4 (PDCD4) mRNA, and its down-regulation leads to 5-FU resistance. When the PDCD4 expression increases, the function of miR-320a is attenuated in a negative feedback loop [11]. mir-301 is another oncogenic miRNA known as an invasion and metastasis-related oncogene that is highly expressed only in pancreatic cancer parallel with cox-2 and MMP-2 proteins which can be a specific pancreatic cancer marker [34]. mir-301 has the potential to be a new target for the prognosis, early diagnosis, and treatment of pancreatic cancer [34]. The cancer was diagnosed in the later phases however, miRNAs can open new windows to a better future [17].

In general, targeting oncogenic miRNAs is a goal of medical strategies. miRNA antagonists are introduced as single-stranded antisense oligodeoxynucleotides (ASO) that target the oncogenic miRNAs, show the anti-cancer effect, and can protect miRNAs from nucleases with high stability and affinity. They can decrease tumor dissemination by inhibiting the function of the oncogenic miRNAs. Thus, providing the basis for integrating miRNAs into pancreatic cancer therapy [11].

REGULATION OF MIRNAS BY NATURE-DERIVED COMPONENTS IN PANCREATIC CANCER THERAPY

Concerning resistance or non-eligibility of pancreatic cancer for various common cancer treatments, like surgery, radiotherapy, and chemotherapy, some researchers instead focused on nature-derived remedies. Natural agents were introduced as promising medical candidates because they do not have any toxic effect on humans. They normalize the level of miRNA in patients, [31] and reduce cancer stem cell resistance

Figure 1 Mechanism of action of A) Oridonin targeted miR-15a by down-regulation of Bmi-1 which inhibit cell proliferation and epithelial to mesenchymal transition in pancreatic ductal adenocarcinoma [39]. B) Genistein targeted miR-223 by up-regulation of F-box and WD-40 domain protein 7 (Fbw7) which suppresses cell growth and induces apoptosis. Genistein targeted miR-27a by up-regulation of ZBTB10 and Sprouty2, blocks NF-????B and Akt signaling pathways which suppresses cell growth, migration, and invasion, and induces apoptosis [60,61]. C) Resveratrol targeted miR-21 by up-regulation of Bcl-2 which increases apoptosis [66]. D) Curcumin targeted miR-22 and miR-199 by down-regulation of Erbb3 and decreases SP1 proteins to inhibit cell proliferation, migration angiogenesis, and metastases [43,59]. E) I3C/DIM targeted miR-221 by up-regulation of PTEN, p27kip1, p57kip2, and PUMA which suppresses cell proliferation and migration of MiaPaCa-2 and Panc-1 pancreatic cancer cells [63]. DIM targeted miR-146 and miR-200 by down-regulates EGFR, IRAK-1, NF-kB, and MTA2 which inhibits cell invasion [64]. DIM targeted miR-200 by down-regulation of ZEB1, slug, vimentin, and E-cadherin which reversal of mesenchymal to epithelial morphology [47]. F) EGCG targeted miR-34a by inhibition of Notch, Bmi1, Ezh2, Suz12, Wnt and sonic hedgehog pathways which selectively blocks self-renewal pathways. EGCG targeted miR-200c by up-regulates the key tumor suppressive miRNAs on pancreatic cancer [65].

Figure 1: Mechanism of action of A) Oridonin targeted miR-15a by down-regulation of Bmi-1 which inhibit cell proliferation and epithelial to mesenchymal transition in pancreatic ductal adenocarcinoma [39]. B) Genistein targeted miR-223 by up-regulation of F-box and WD-40 domain protein 7 (Fbw7) which suppresses cell growth and induces apoptosis. Genistein targeted miR-27a by up-regulation of ZBTB10 and Sprouty2, blocks NF-????B and Akt signaling pathways which suppresses cell growth, migration, and invasion, and induces apoptosis [60,61]. C) Resveratrol targeted miR-21 by up-regulation of Bcl-2 which increases apoptosis [66]. D) Curcumin targeted miR-22 and miR-199 by down-regulation of Erbb3 and decreases SP1 proteins to inhibit cell proliferation, migration angiogenesis, and metastases [43,59]. E) I3C/DIM targeted miR-221 by up-regulation of PTEN, p27kip1, p57kip2, and PUMA which suppresses cell proliferation and migration of MiaPaCa-2 and Panc-1 pancreatic cancer cells [63]. DIM targeted miR-146 and miR-200 by down-regulates EGFR, IRAK-1, NF-kB, and MTA2 which inhibits cell invasion [64]. DIM targeted miR-200 by down-regulation of ZEB1, slug, vimentin, and E-cadherin which reversal of mesenchymal to epithelial morphology [47]. F) EGCG targeted miR-34a by inhibition of Notch, Bmi1, Ezh2, Suz12, Wnt and sonic hedgehog pathways which selectively blocks self-renewal pathways. EGCG targeted miR-200c by up-regulates the key tumor suppressive miRNAs on pancreatic cancer [65].

to traditional agents. Numerous analyses confirm that consuming natural agents regulate miRNA expression [32]. Figure 1 presents the targets, mechanism of action, and consequence of several effective nature-derived components such as oridonin, curcumin, isoflavone, I3C, DIM, EGCG, and resveratrol against pancreatic cancer

One of the natural agents is oridonin, which is a natural terpenoid in traditional Chinese herbal medicine [33]. It has some antitumor effects, such as, the promotion of apoptosis, as well as cell growth and invasion inhibition [34]. According to a microarray profile analysis in BxPC-3 human pancreatic cancer cells, one hundred and five miRNAs are differentially expressed following oridonin treatment, confirming the potential of oridonin as a candidate in future research on tumor treatments [35,36]. The effect of oridonin has not been evaluated in any cancers in clinical trials. For filling this information gap, conducting a clinical trial to gain evidence of the oridonin effect on cancer treatment is highly recommended.

Curcumin (diferuloylmethane) is another natural agent derived from turmeric. It boosts with antioxidant, antiproliferative, pro-apoptotic, anti-inflammatory, and tumor inhibitory properties and control of the cell cycle by regulating signaling pathways such as NF-κB, and the expression of their target genes [37,38]. For example, in human pancreatic cells, curcumin up-regulates miR-22, the tumor suppressor gene while it down-regulates the miR-199a, an oncogene [39]; Despite its promising mechanism of action, its administration is limited because of its weak bioavailability [31].

Soy Isoflavones like genistein, daidzein, and glycitein are other natural agents that are derived from soybean. Genistein inhibits tumor progression, cancer cell proliferation, invasion, and metastasis through oncomir degradation [40-43]. The miR-200 and let-7 families are down-regulated in gemcitabineresistant pancreatic cancer cells with EMT conditions which lead to pancreatic cancer aggressiveness [43]. Isoflavones increase miR-200 expression and subsequently, decrease vimentin expression which reverses the EMT phenotype. As a result, it also up-regulates let-7 and inhibits cancer cell growth.

Dimer diindolylmethane (DIM) and Indole-3-carbinol (I3C) are driven by glucosinolates that are in the Cruciferae plant family. In fact, in vivo, I3C dimerizes with DIM. They restrict cancer cell proliferation and metastasis by EMT-related miRNA gene regulation. They affect both tumor suppressors and oncogenic miRNAs. It has been observed that in lung cancer, I3C normalizes miRNA abnormalities [5]. For example, it down-regulates miR21, miR-31, miR-130a, miR-146b, and miR-377. Moreover, I3C inhibits PTEN, PDCD4, and RECK as miR-21 target genes while in pancreatic cancer, DIM is EMT reversing. DIM also up-regulates miR-200 and let-7 families in gemcitabine resistant cells. These miRNAs usually have been lost in many cancers including pancreatic cancer. Conversely, DIM down-regulates vimentin and slug in mesenchymal to an epithelial phenotype. Therefore, it is a potent nature-derived therapeutic component against pancreatic cancer [6].

Additionally, it has been shown that DIM elevated the level of miR-34a in prostate cancer via epigenetic regulation. The miR-34a re-expression resulted in the androgen receptor’s (AR) down-regulation [44]. These findings demonstrate the positive impacts of DIM on the activation of tumor-suppressive miRNAs. It suggests a potential therapeutic strategy for the treatment of human prostate cancer. More research on other malignancies, including pancreatic cancer, will shed light on the therapeutic agent’s significance in preventing cancer.

EGCG (Epigallocatechin-3-gallate) is a polyphenol derived from natural green tea with anticancer characteristics through effects on miRNAs [45-47]. EGCG down-regulates 48 miRNAs and up-regulates 13 miRNAs such as miR-16 in HepG2 human hepatic cancer cells. The latter leads to apoptosis through a decrease of Bcl-2 in this cell line [47]. Based on such data, EGCG can inhibit cancer growth through miRNA regulation.

Resveratrol is a natural phytoalexin that can be found in some plants such as mulberries, grapes, and peanuts. It has anticancer effects by miRNAs regulation that leads to cancer cells growth inhibition and apoptosis induction [48, 49]. CAY10512 is a synthetic analog of resveratrol that can affect miR-146a expression [50]. Based on this research, resveratrol plays a role in changing physiological characteristics of cells by miRNA expression regulation and their subsequent signal transduction [35]. Therefore, this nature-derived compound as a controller of cell proliferation, resveratrol can be considered for wider anticancer investigations.

THE SELECTED NATURE-DERIVED COMPONENTS IN THE CLINICAL TRIALS

This study focused on pancreatic cancer in clinical trials, including a thorough literature assessment on the chosen naturederived components against malignancies (Table 1). The findings of this investigation revealed that while other cellular processes like signal transduction have been taken into consideration, the effects of oridonin and curcumin on miRNA levels have not been explored in any clinical trials with respect to malignancies. For example, in the Phase 2 clinical trial (NCT00094445), 21 pancreatic cancer patients received curcumin orally to assess its biological effects. The authors reported no toxicities nor side effects, and a down-regulation of NF-κB, cyclooxygenase-2, and phosphorylated STAT3 in mononuclear immune cells of peripheral blood obtained from patients [51]. Most of the patient baseline levels of NF-κB, cyclooxygenase-2, and phosphorylated STAT3 were significantly higher than their counterparts in healthy controls [51]. In another investigation, the curcumin effect was evaluated in cancer patients who were treated with gemcitabine. This study is in phase 2 clinical trial (NCT00192842). In another trial which is a recruiting phase III (NCT00486460), the effect of three drugs combination of gemcitabine, curcumin and Celebrex have been evaluated in patients suffering from advanced or inoperable pancreatic cancers. There are no released results from this study, but in theory, curcumin can be considered a successful candidate alone or in combination with conventional drugs.

Since resveratrol inhibits miR-21 and acts as an antiapoptotic agent of BCL-2. Specifically, it could potentially reduce the growth of cancerous cells in the pancreas and bladder. The effectiveness of resveratrol as an anti-cancer drug has been assessed in numerous clinical trials on a wide range of cancer

Table 1: Clinical trials evaluating the effects of curcumin, isoflavone, I3C, DIM, EGCG, and resveratrol on various human cancers.

Natural component Cancer type Trial identifier Phase
Curcumin Breast NCT01042938 2
Pancreatic NCT00094445 2
Pancreatic NCT00192842 2
Colorectal NCT01333917 1
Colorectal NCT00027495 1
Uterine cervical NCT01035580 1
Multiple myeloma NCT00113841 N.A
Breast NCT01975363 N.A
Colorectal NCT01859858 1
Rectal NCT00745134 2
Colon NCT01294072 1
Prostate NCT01917890 N.A
Head and neck NCT01160302 0
Colon NCT01490996 1/2
Prostate NCT02095717 2
Endometrial NCT02017353 2
Prostate NCT02064673 2
Familial adenomatous Polyposis NCT00641147 N.A
Familial adenomatous Polyposis NCT00927485 N.A
Colorectal NCT01948661 2
Resveratrol Colon NCT00256334 1
Colorectal NCT00433576 1
Solid tumors NCT00098969 1
Neuroendocrine NCT01476592 N.A
Genistein Breast NCT00244933 2
Bladder NCT00118040 2
Breast NCT00099008 1
Prostate NCT00584532 2/3
Pancreatic NCT00376948 2
Prostate NCT00269555 N.A
Prostate NCT00499408 2
Prostate NCT00078923 2
Colon cancer, rectal cancer, and colorectal cancer NCT01985763 1/2
Prostate Adenocarcinoma NCT01325311 2
Adenocarcinoma of the prostate and recurrent prostate cancer NCT01126879 2
Non-small cell lung cancer NCT01628471 1/2
Kidney Cancer and Melanoma (Skin) NCT00276835 1
Pancreatic NCT01182246 1/2
EGCG Adenocarcinoma of the prostate NCT00459407 1
Barrett esophagus NCT00233935 1
Lung cancer and tobacco use disorder NCT00573885 2
Cervical cancer, cervical intraepithelial neoplasia grade 1, and human papilloma virus infection NCT00303823 2
Prostate NCT01105338 2/3
Estrogen receptor-negative breast cancer and progesterone receptornegative breast cancer NCT00516243 1
Multiple myeloma, plasma cell neoplasm, and precancerous condition NCT00942422 2
Prostatic hyperplasia NCT00596011 2
Adenocarcinoma of the colon NCT01606124 2
Precancerous condition prostate cancer NCT00253643 N.A
Breast NCT00917735 2
Colorectal NCT01360320 2
Breast NCT00949923 N.A
I3C Prostate NCT00607932 N.A
Breast NCT00033345 1
Unspecified adult solid tumor NCT00100958 1
DIM Prostate NCT00450229 1
Prostate NCT00305747 1
Cervical cancer and precancerous condition NCT00462813 3
Prostate NCT00888654 2
Breast NCT01391689 2/3

and malignancies including colon cancer prevention by blocking the Wnt pathway (an important signaling pathway in vitro and in animal colon cancer). Currently, 18 studies have assessed colon cancer, liver cancer, neuroendocrine tumors, neoplasms, colorectal adenocarcinoma, unspecified adult solid tumor, breast cancer, multiple myeloma, lymphangioleiomyomatosis, but no trial has studied the effect of resveratrol on miRNAs in pancreatic cancer or other cancers. We thus highly-recommend designing a trial in this area.

No studies have been found for the evaluation of soy isoflavones’ effects on pancreatic cancer, but a couple of studies on other cancers may prove useful. For example, the effects of soy isoflavone on prostate-specific antigen (PSA), hormone levels, gene expression, and apoptosis were evaluated in men suffering from localized prostate cancer in a clinical trial (NCT00255125). The gene expression data showed twelve genes related to the control of the cell cycle and nine genes related to apoptosis being down-regulated in the treatment group compared to the placebo control group [52]. Designing a novel study for the assessment of isoflavone’s effect on pancreatic cancer patients can be useful.

Similarly, there are no studies for assessing the effects of BioResponse 3,3′-diindolylmethane (BR-DIM) in pancreatic cancer. However, 52 studies were found evaluating this natural component’s effects on different cancers. For example, in the phase II clinical trial (NCT00888654), the effect of DIM treatment on castration-resistant prostate cancer (CRPC) was assessed. The result showed BR-DIM down-regulated the wild-type AR in vitro and in vivo. AR can induce epithelial-to-mesenchymal transition (MET) phenotypes and maintain stem cell characteristics; both phenotypes lead to enzalutamide resistance. The enzalutamide resistance occurs partly because the AR, Lin28B, and EZH2 are dysregulated via the re-expression of miR-34a, miR-320, miR27b, and let-7 in human prostate cancer (PCa). The final analysis proved that BR-DIM was well-tolerated in these clinical trials, and in 93% of patients, prostatic DIM levels were detectable. The blocking effects of BR-DIM on the AR, its target genes, and PSA were detectable, and a modest efficacy was observed. As a result, BR-DIM is a potential medical candidate to delay or prevent cancer development [53] and more studies on the evaluation of its effects on pancreatic cancer in trials is necessary.

The effect of EGCG on miRNAs in cancers has not been evaluated in any trial and only one study has evaluated the effect of EGCG in pancreatic adenocarcinoma and unresectable pancreatic carcinoma. The status of this phase I clinical trial (NCT02336087) is active, not recruiting and no data has been released yet. Forty-one studies were found for the evaluation of EGCG in other cancers. As obesity has an important role in cancer progression, the control of obesity is effective in cancer treatment. For example, in a trial (NCT00596011), the effect of EGCG was assessed on obese men suffering from prostate cancer. The results demonstrated that EGCG was not effective in reducing body mass index and subsequently it was not effective in cancer treatment [54]. This trial failed to meet the endpoints but more studies in this area are needed.

As there are no trials concerning the effect of resveratrol on miRNAs in pancreatic or any other cancers, we present an analysis of its effects on other intracellular compounds. In a phase I pilot study on eight patients suffering from colon cancer (NCT00256334), the efficacy of low dose resveratrol and resveratrol in the form of freeze-dried grape powder (GP) was evaluated on Wnt signal transduction in the colon. The result of this trial shows GP in combination with various bioactive components inhibits the Wnt pathway in the normal colonic mucosa, but not cancerous ones. More studies on supplements containing whole grapes or GP as a potent colon cancer drug are warranted [55].

CONCLUSIONS

According to our earlier studies and the literature analysis, miRNAs are effective cancer therapy targets because when one of them is altered, a network of genes is subsequently affected. Due to CSCs as well as EMT phenotypes, the modified pattern of miRNA expression is associated with tumor initiation, progression, invasion, and metastasis. The deregulation of miRNA expression modifies the biological activity of the processes. miRNAs are effective diagnostic, prognostic, and therapeutic agents due to their sequence specificity on the target mRNA and their ability to prevent mRNAs translation in the first place.

Natural substances provide new possibilities for therapies since they simultaneously target several miRNAs, which in turn regulate multiple targets. Curcumin, isoflavones, I3C, DIM, EGCG, and resveratrol are examples of natural substances that modulate miRNAs to prevent the development of cancer cells, trigger apoptosis, and turn EMT into MET phenotypes. They can therefore be used as chemo-preventive drugs in lieu of traditional treatments. The natural substances remove CSCs or EMT phenotypic cells, which are responsible for tumor recurrence and resistance to several conventional treatments. As a result, natural remedies increase the overall survival of cancer patients [31]. Few clinical studies have been conducted to evaluate natural therapeutic agents for pancreatic cancer; therefore, new clinical trials should be planned.

REFERENCES

1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019; 69: 7-34.

2. Hussain SP. Pancreatic cancer: current progress and future challenges. Int J Biol Sci. 2016; 12: 270.

3. Chandra Gupta S, Nandan Tripathi Y. Potential of long non?coding RNAs in cancer patients: from biomarkers to therapeutic targets. Int J Cancer. 2017; 140: 1955-1967.

4. Duguang L, Jin H, Xiaowei Q, Peng X, Xiaodong W, Zhennan L, et al. The involvement of lncRNAs in the development and progression of pancreatic cancer. Cancer Biol Ther. 2017; 18: 927-936.

5. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993; 75: 843-854.

6. Chen S, Xu J, Su Y, Hua L, Feng C, Lin Z, et al. MicroRNA-145 suppresses epithelial to mesenchymal transition in pancreatic cancer cells by inhibiting TGF-β signaling pathway. J Cancer. 2020; 11: 2716.

7. Khan S, Ebeling MC, Zaman MS, Sikander M, Yallapu MM, Chauhan N, et al. MicroRNA-145 targets MUC13 and suppresses growth and invasion of pancreatic cancer. Oncotarget. 2014; 5: 7599.

8. Han T, Yi XP, Liu B, Ke MJ, Li YX. MicroRNA-145 suppresses cell proliferation, invasion and migration in pancreatic cancer cells by targeting NEDD9. Mol Med Rep. 2015; 11: 4115-4120.

9. An N, Zheng B. MiR-203a-3p inhibits pancreatic cancer cell proliferation, EMT, and apoptosis by regulating SLUG. Technol Cancer Res Treat. 2020; 19: 1533033819898729.

10.Wang W, Zhao L, Wei X, Wang L, Liu S, Yang Y, et al. MicroRNA-320a promotes 5-FU resistance in human pancreatic cancer cells. Scientific reports. 2016, 6: 1-11.

11. Chu X, Wei D, Liu X, Long D, Tian X, Yang Y. MicroRNAs as potential therapeutic targets for pancreatic cancer. Chin Med J (Engl). 2022; 135: 4-10.

12. Zhan T, Zhu Q, Han Z, Tan J, Liu M, Liu W, et al. miR-455-3p functions as a tumor suppressor by restraining Wnt/β-catenin signaling via TAZ in pancreatic cancer. Chin Med J (Engl). 2020; 12: 1483.

13. Hu W, Liu Q, Pan J, Sui Z. miR-373-3p enhances the chemosensitivity of gemcitabine through cell cycle pathway by targeting CCND2 in pancreatic carcinoma cells. Biomed Pharmacother. 2018; 105: 887- 898.

14. Liu W, Lu Y, Zhang D, Shi L, Zu G, Yan H, et al. MicroRNA-708 inhibits the proliferation and chemoresistance of pancreatic cancer cells. Biocell. 2020; 44: 73.

15.Tang Y, Tang Y, Cheng YS. miR-34a inhibits pancreatic cancer progression through Snail1-mediated epithelial–mesenchymal transition and the Notch signaling pathway. Sci Rep. 2017; 7: 1-11.

16. Zhang QA, Yang XH, Chen D, Yan X, Jing FC, Liu HQ, et al. miR-34 increases in vitro PANC-1 cell sensitivity to gemcitabine via targeting Slug/PUMA. Cancer Biomarkers. 2018; 21: 755-762.

17. Lv Y, Huang S. Role of non coding RNA in pancreatic cancer. Oncol let. 2019l; 18: 3963-3973.

18. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A. 2002; 99: 15524-15529.

19. Kalhori MR, Soleimani M, Irani S. MiR-16 induces G1 cell cycle arrest and apoptosis in acute myeloid leukemia cells. Gene Reports. 2019; 15: 100399.

20. Karamitopoulou E, Haemmig S, Baumgartner U, Schlup C, Wartenberg M, Vassella E. MicroRNA dysregulation in the tumor microenvironment influences the phenotype of pancreatic cancer. Mod pathol. 2017; 30: 1116-1125.

21. Arab A, Karimipoor M, Irani S, Kiani A, Zeinali S, Tafsiri E, et al. The Evaluation of miR-21 Level in Lung Tissue and Plasma of Nsclc Patients. Basic & Clinical Cancer Research. 2017; 9: 13-21.

22. Zhang L, Yao J, Li W, Zhang C. Micro-RNA-21 regulates cancerassociated fibroblast-mediated drug resistance in pancreatic cancer. Oncol res. 2018; 26: 827.

23. Wei X, Wang W, Wang L, Zhang Y, Zhang X, Chen M, et al. Micro RNA?21 induces 5?fluorouracil resistance in human pancreatic cancer cells by regulating PTEN and PDCD4 . Cancer med. 2016; 5: 693-702.

24. Zhao Q, Chen S, Zhu Z, Yu L, Ren Y, Jiang M, et al. miR-21 promotes EGF-induced pancreatic cancer cell proliferation by targeting Spry2. Cell Death Dis. 2018; 9:1157.

25. Sun J, Jiang Z, Li Y, Wang K, Chen X, Liu G. Downregulation of miR21 inhibits the malignant phenotype of pancreatic cancer cells by targeting VHL. OncoTargets Ther. 2019; 12: 7215-7226.

26. Negoi I, Hostiuc S, Sartelli M, Negoi RI, Beuran M. MicroRNA-21 as a prognostic biomarker in patients with pancreatic cancer–a systematic review and meta-analysis. Am J Surg. 2017; 214: 515-524.

27. Wang P, Zhu CF, Ma MZ, Chen G, Song M, Zeng ZL, et al. Micro-RNA-155 is induced by K-Ras oncogenic signal and promotes ROS stress in pancreatic cancer. Oncotarget. 2015; 6: 21148-58.

28. Huang C, Li H, Wu W, Jiang T, Qiu Z. Regulation of miR-155 affects pancreatic cancer cell invasiveness and migration by modulating the STAT3 signaling pathway through SOCS1. Oncology Rep. 2013; 30: 1223-1230.

29. Wang J, Guo J, Fan H. MiR-155 regulates the proliferation and apoptosis of pancreatic cancer cells through targeting SOCS3. Eur Rev Med Pharmacol Sci. 2019; 23: 5168-5175.

30. Taheri Z, Aghdaei HA, Irani S, Modarressi MH, Noormohammadi Z. Clinical correlation of miR-200c/141 cluster DNA methylation and miR-141 expression with the clinicopathological features of colorectal primary lesions/tumors. Rep Biochem Mol Biol. 2019; 8: 208.

31. Sethi S, Li Y, H Sarkar F. Regulating miRNA by natural agents as a new strategy for cancer treatment. Curr drug targets. 2013; 14: 1167-1174.

32.Dolganiuc A, Petrasek J, Kodys K, Catalano D, Mandrekar P, Velayudham A, et al. MicroRNA expression profile in Lieber?DeCarlidiet?induced alcoholic and methionine choline deficient diet?induced nonalcoholic steatohepatitis models in mice. Alcohol Clin Exp Res. 2009; 33: 1704-1710.

33. Li X, Zhang CT, Ma W, Xie X, Huang Q. Oridonin: a review of its pharmacology, pharmacokinetics and toxicity. Front Pharmacol. 2021; 12: 645824.

34. Wang S, Zhong Z, Wan J, Tan W, Wu G, Chen M, et al. Oridonin induces apoptosis, inhibits migration and invasion on highly-metastatic human breast cancer cells. Am J Chin Med. 2013; 41: 177-196.

35. Gui Z, Li S, Liu X, Xu B, Xu J. Oridonin alters the expression profiles of microRNAs in BxPC-3 human pancreatic cancer cells. BMC complementary and alternative medicine. 2015; 15: 1-10.

36. Qadir MI, Faheem A. miRNA: A diagnostic and therapeutic tool for pancreatic cancer. Crit Rev Eukaryot Gene Expr. 2017; 27: 197-204.

37. Mukhopadhyay A, Bueso-Ramos C, Chatterjee D, Pantazis P, Aggarwal BB. Curcumin downregulates cell survival mechanisms in human prostate cancer cell lines. Oncogene. 2001; 20: 7597-7609.

38. Shao ZM, Shen ZZ, Liu CH, Sartippour MR, Go VL, Heber D, et al. Curcumin exerts multiple suppressive effects on human breast carcinoma cells. Int J cancer. 2002; 98: 234-240.

39. Sun M, Estrov Z, Ji Y, Coombes KR, Harris DH, Kurzrock R. Curcumin (diferuloylmethane) alters the expression profiles of microRNAs in human pancreatic cancer cells. Mol cancer Ther. 2008; 7: 464-473.

40. Li Y, Sarkar FH. Down-regulation of invasion and angiogenesis-related genes identified by cDNA microarray analysis of PC3 prostate cancer cells treated with genistein. Cancer Lett. 2002; 186: 157-164.

41. Dixon RA, Ferreira D. Genistein. Phytochemistry. 2002; 60: 205-11.

42. Barnes S. The chemopreventive properties of soy isoflavonoids in animal models of breast cancer. Breast cancer Res Treat. 1997; 46: 169-179.

43. Li Y, VandenBoom TG, Kong D, Wang Z, Ali S, Philip PA, et al. Upregulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res. 2009; 69: 6704-6712.

44. Paik WH, Kim HR, Park JK, Song BJ, Lee SH, Hwang JH. Chemosensitivity induced by down-regulation of microRNA-21 in gemcitabine-resistant pancreatic cancer cells by indole-3-carbinol. Anticancer Res. 2013; 33: 1473-1481.

45. Katiyar SK, Afaq F, Perez A, Mukhtar H. Green tea polyphenol (–)-epigallocatechin-3-gallate treatment of human skin inhibits ultraviolet radiation-induced oxidative stress. Carcinogenesis. 2001; 22: 287-294.

46. Mukhtar H, Ahmad N. Green tea in chemoprevention of cancer. Toxicol Sci.1999; 52: 111-117.

47. Tsang WP, Kwok TT. Epigallocatechin gallate up-regulation of miR16 and induction of apoptosis in human cancer cells. J Nutr Biochem. 2010; 21: 140-146.

48. Kundu JK, Surh YJ. Cancer chemopreventive and therapeutic potential of resveratrol: mechanistic perspectives. Cancer Lett. 2008; 269: 243- 261.

49. Whyte L, Huang YY, Torres K, Mehta RG. Molecular mechanisms of resveratrol action in lung cancer cells using dual protein and microarray analyses. Cancer Res. 2007; 67: 12007-12017.

50.Lukiw WJ, Zhao Y, Cui JG. An NF-κB-sensitive micro RNA-146amediated inflammatory circuit in Alzheimer disease and in stressed human brain cells. J Biol Chem. 2008; 283: 31315-31322.

51. Dhillon N, Aggarwal BB, Newman RA, Wolff RA, Kunnumakkara AB, Abbruzzese JL, et al. Phase II trial of curcumin in patients with advanced pancreatic cancer. Clin Cancer Res. 2008; 14: 4491-4499.

52. Hamilton-Reeves JM, Banerjee S, Banerjee SK, Holzbeierlein JM, Thrasher JB, Kambhampati S, et al. Short-term soy isoflavone intervention in patients with localized prostate cancer: a randomized, double-blind, placebo-controlled trial. PloS one. 2013; 8: e68331.

53. Li Y, Sarkar FH. Role of BioResponse 3, 3′-diindolylmethane in the treatment of human prostate cancer: Clinical experience. Med Princ Pract. 2016; 25: 11-17.

54. Kumar NB, Patel R, Pow-Sang J, Spiess PE, Salup R, Williams CR, et al. Long-term supplementation of decaffeinated green tea extract does not modify body weight or abdominal obesity in a randomized trial of men at high risk for prostate cancer. Oncotarget. 2017; 8: 99093.

55. Nguyen AV, Martinez M, Stamos MJ, Moyer MP, Planutis K, Hope C, et al. Results of a phase I pilot clinical trial examining the effect of plantderived resveratrol and grape powder on Wnt pathway target gene expression in colonic mucosa and colon cancer. Cancer Manag Res. 2009; 1: 25-37.

56. Sreenivasan S, Thirumalai K, Danda R, Krishnakumar S. Effect of curcumin on miRNA expression in human Y79 retinoblastoma cells. Curr Eye Res. 2012; 37: 421-428.

57. Ma J, Cheng L, Liu H, Zhang J, Shi Y, Zeng F, et al. Genistein downregulates miR-223 expression in pancreatic cancer cells. Curr Drug Targets. 2013; 14: 1150-1156.

58. Xia J, Cheng L, Mei C, Ma J, Shi Y, Zeng F, et al. Genistein inhibits cell growth and invasion through regulation of miR-27a in pancreatic cancer cells. Curr Pharm Des. 2014; 20: 5348-5353.

59. Sarkar S, Dubaybo H, Ali S, Goncalves P, Kollepara SL, Sethi S, et al. Down-regulation of miR-221 inhibits proliferation of pancreatic cancer cells through up-regulation of PTEN, p27kip1, p57kip2, and PUMA. Am J Cancer Res. 2013; 3: 465-77.

60. Li Y, VandenBoom TG, Wang Z, Kong D, Ali S, Philip PA, et al. miR-146a suppresses invasion of pancreatic cancer cells. Cancer Res. 2010; 70: 1486-1495.

61.Tang SN, Fu J, Nall D, Rodova M, Shankar S, Srivastava RK. Inhibition of sonic hedgehog pathway and pluripotency maintaining factors regulate human pancreatic cancer stem cell characteristics. Int J Cancer. 2012; 131: 30-40.

62. Liu P, Liang H, Xia Q, Li P, Kong H, Lei P, et al. Resveratrol induces apoptosis of pancreatic cancers cells by inhibiting miR-21 regulation of BCL-2 expression. Clin Transl Oncol. 2013; 15: 741-746.

Taheri Z, Irani S, Dashti MG, Esfahani NB, Heidari A, et al. (2023) microRNAs as Therapeutic Targets in Diagnosis, Prognosis, and Treatment of Pancreatic Cancer: A Mini Review. Ann Food Process Preserv 7(1): 1035.

Received : 25 Jan 2023
Accepted : 09 Feb 2023
Published : 10 Feb 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
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 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