Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2021 Jun 15;11(6):2386–2400.

Basic approaches, challenges and opportunities for the discovery of small molecule anti-tumor drugs

Yu-Shui Ma 1,2,3,*, Ji-Bin Liu 2,*, Xiao-Li Yang 3, Rui Xin 3, Yi Shi 1,2, Dan-Dan Zhang 3, Hui-Min Wang 3, Pei-Yao Wang 3, Qin-Lu Lin 1, Wen Li 1, Da Fu 1,3
PMCID: PMC8263657  PMID: 34249406

Abstract

Chemotherapy is one of the main treatments for cancer, especially for advanced cancer patients. In the past decade, significant progress has been made with the research into the molecular mechanisms of cancer cells and the precision medicine. The treatment on cancer patients has gradually changed from cytotoxic chemotherapy to precise treatment strategy. Research into anticancer drugs has also changed from killing effects on all cells to targeting drugs for target genes. Besides, researchers have developed the understanding of the abnormal physiological function, related genomics, epigenetics, and proteomics of cancer cells with cancer genome sequencing, epigenetic research, and proteomic research. These technologies and related research have accelerated the development of related cancer drugs. In this review, we summarize the research progress of anticancer drugs, the current challenges, and future opportunities.

Keywords: Drug therapy, cancer, inhibitor, comprehensive utilization


In the 1940s, nitrogen mustard was found to cure malignant lymphoma in humans [1] which has boosted researchers’ confidence in curing cancer. With the rapid development of biology, people have researched the subjects including biochemistry, immunology, and therapeutics [2-4], leading the cognition of tumors to a genetic level [5-7]. The clinical application of various antitumor drugs has continually prompted researchers to explore many new antitumor drugs [8-10].

Small molecule drugs mainly refer to chemical synthetic drugs with molecular weight less than 1000, of which structure has good spatial dispersion and leads to their high drug efficiency and pharmacokinetic properties [11]. As a result, the market started to focus on those drugs. Small molecule anticancer drugs, usually signal transduction inhibitors, can block the signal transduction pathways for tumor growth and proliferation, then to cure the tumor [12] such as, Novartis Gleevec’s for the treatment of chronic myeloid leukemia and gastrointestinal stromal tumors and AstraZeneca’s treatment of non-small cell lung cancer with epidermal growth factor receptor (EGFR) as the target. Small molecule drugs have already had the advantages of wide use and mature theory.

Although small molecule drugs have achieved encouraging results, there are still many challenges. Disobedience of the rational drug use would increase the side effects and drug tolerance, leading to poor treatment effect [13]. Secondly, small molecule drug monotherapy, especially protease inhibitors, cancer cells are prone to cause drug-resistant mutations in about 2 weeks [14]. In addition, small molecule drugs are prone to generate multi drug resistance sites. In a word, the small molecule drugs are in the ascendant, and new drugs are constantly emerging, which has made the anti-cancer and anti-tumor drug treatment essential [15]. With the continuous development of such drugs with low drug resistance, high efficacy and few side effects, it is believed that in the near future, there will be significance breakthrough in the treatment of cancer.

Nowadays, novel and effective antitumor drugs is urgently needed. The basic approaches, challenges, and opportunities for the discovery of antineoplastic agents are summarized below.

Drug target selection

Choosing the right target needs to balance benefits and risks, which is one of the most critical problems [19-21]. Wrong drug target is a costly waste of drug research and development [22-24]. In general, the selection of targets depends on a detailed understanding of the molecular mechanism (Figure 1). After the target activity is regulated in pharmacology (including activation or inhibition), it can achieve antitumor effects both in vitro cell system and in vivo, and has selectivity for cancer cells with few side effects [25-27].

Figure 1.

Figure 1

Discovery and development from gene to drug. Small molecules that act on new molecular targets represent therapeutic dependencies and vulnerabilities. There are four main steps of cancer drug discovery: target selection and validation, chemical hit and lead generation, lead optimization to select a clinical candidate, and biology-led clinical trials.

Naturally-derived drugs

Naturally-derived drugs kill tumor cells by inhibiting proliferation and inducing apoptosis aiming at metabolic heterogeneity; the drugs also act on tumor cells in indirect manners, such as immune regulation and epithelial-mesenchymal transition (EMT) inhibition of metastasis (Figure 2). Therefore, researchers have focused on identifying natural products to use as drugs for treating cancer [29].

Figure 2.

Figure 2

Potential drug targets for targeted therapy. Common proteins upregulated in the majority of tumors were classified in the four functional categories: proteins involved in extracellular matrix (ECM), immune response, cell cycle/DNA replication, and metabolism.

At present, more than 2,000 plants have been screened for anticancer activity in China, 190 have shown anticancer activity in animal experiments [30]. Paclitaxel is an alkaloid compound isolated and purified from the bark of the gymnosperm yew [31]. When treated with paclitaxel, the cells would accumulate microtubules interfere with various functions of the cells, such as blocking the normal cell division [32]. In Phase II-III clinical studies, paclitaxel is mainly used for ovarian cancer [33] and breast cancer [34]; it has also shown to have some beneficial effects against lung cancer, colorectal cancer and melanoma [35-38]. Another plant called vinorelbine is a cell cycle-specific drug with semi-synthetic compound [39]; the plant navelbine could stop mitosis in the metaphase by blocking tubulin polymerization to form microtubules and induce tubule formation disorders [40,41].

Synthetic antitumor drugs

Chemical antitumor drug refers to cytotoxic drug and acts on the chemical structure of DNA [42], which plays an irreplaceable role in treating cancer; These drugs are mainly alkylating agents, antimetabolites, antitumor antibiotics, platinum complexes, and targeted drugs [43]. However, while these drugs kill cancer cells, normal cells would also receive detrimental effects [45]. Besides, the significant drug resistance and allergic reactions have also limited their long-term clinical applications [46-48]. Therefore, chemical antitumor drugs with low toxicity and high efficiency are an urgent issue for pharmaceutical companies.

Alkylating agents were one of the earliest classes of drugs for cancer treatment are mainly used for chronic lymphocytic leukemia and malignant lymphoma [49], which could generate a carbocation or other compound with an electrophilic group in vivo [50]. These reactive electrophilic species can covalently combine with electron-rich groups (e.g., amino, sulfhydryl, hydroxyl, carboxyl, and phosphate) in biological macromolecules (e.g., DNA, RNA, and enzymes) in cells [51-53]. This combination would cause loss of biological macromolecule activity or cleavage of DNA molecules, resulting in death of tumor cell and antitumor activity [54-58].

Antimetabolites are a class of drugs that affect the biosynthesis of nucleic acids [59], which chemical structure is similar to what nucleic acid metabolism required. Antimetabolitescan prevent cell division and proliferation by specifically interfering with the metabolism of nucleic acids including methotrexate, 6-mercaptopurine, 5-fluorouracil (5-FU), and cytarabine [60,61]. Deoxyfluorouridine is a prodrug of 5-FU [62], which could be decomposed into 5-FU by pyrimidine nucleoside phosphorylase after entering the body in order to kill the tumor; it is used clinically to treat breast and digestive tract tumors clinical [63]. Is a congener of cytarabine [64], its one nucleotide can allow intrusion, and the second nucleotide can inhibit the multimerization process after breaking DNA, forming a mask chain break [65]. Difluoro deoxy cytarabinealso inhibits nucleotide reductase and inhibits DNA synthesis, which means it is one of the first choices in treating non-small cell lung cancer (NSCLC) and pancreatic tumors [66]. Antitumor antibiotics, containing dimethoxy daunorubicin, which has high fat solubility and a strong anti-leukemia effect, are extracted from a class of microbial culture fluids that interfere with transcription by directly disrupting DNA or embedding DNA [67]. Puromycinis a semi-synthetic antibiotic with a tetrahydropyran at the 4-position of doxorubicin [68] which can inhibit DNA replication and transcription, and block the cell cycle in the G2 phase; it has a wide antitumor spectrum and can inhibit tumor metastasis [69]. The cardiotoxicity of puromycin is no higher than epirubicin, it’s side effect of hair loss is significantly lower than other anthracyclines during the clinical treatment of breast cancer and lymphoma [70].

The anticancer mechanism of platinum antitumor drugs can be divided into four steps: transmembrane transport, hydration dissociation, targeted migration, and action on DNA, which may cause DNA replication disorders, thereby inhibiting the division of cancer cells [71-73]. Preclinical studies have shown significant inhibitory effects on colorectal tumor cell lines and cisplatin-resistant cell lines, and significant synergistic effects with 5-FU [74-76]. Nidaplatin is more effective than isodose cisplatin in the head and neck, testis, lung, esophagus, bladder, ovary, and cervical tumors, while digestive tract reaction and nephrotoxicity are mild [77-79].

Molecularly targeted drugs are often targeted at key enzymes in cell signaling pathways involved in tumor cell differentiation and proliferation, also screen for low-toxic, highly potent, and specific small molecule compounds that selectively act on specific targets [80-82]. It was first used clinically in an antitumor small molecule compound with a single kinase target, which easily produces drug resistance with a narrow therapeutic range [83]. Most solid tumors are multi-link and multi-target. Multinomial integration analysis that are essential tools for stratifying patients according to risk factors provide insights to use more targeted and individualized therapeutics (Figure 3). Blocking a certain target or receptor does not need to block the signal transduction of all cells, so multi-kinase targeting represent a new development direction of tumor-targeted therapeutic [84], including targeted drugs that inhibit tumor angiogenesis, protein tyrosine kinase inhibitors, and mammalian target of rapamycin (mTOR) inhibitors [85].

Figure 3.

Figure 3

A road map to personalizing targeted cancer therapies using multinomial integration analysis. Multinomial integration analysis approach has been used for personalized targeted therapeutics in a genome profiled patient cohort. Mass spectrometry (MS)-based proteomics can measure global protein abundance and post-translational modifications to provide additional biological insights, which may not be deciphered by genomic analysis alone. The combination of sequencing and MS provides a more comprehensive picture linking cancer genotype to phenotype through functional proteomics and signaling networks. Integrated analyses of genomic, transcriptomic, proteomic, and phosphoproteomic data from tumor and matched non-tumor liver tissues revealed the connection and discordance among multi-omics and alterations in critical signaling and metabolic pathway. Thus, tumor genome analysis for mutation in a cancer-specific gene as a biomarker results in a better outcome in clinical trials.

Targeted drugs that inhibit tumor angiogenesis include inhibit vascular endothelial growth factor (VEGF) such as bevacizumab for the treatment of NSCLC, panitumumab, cetuximab, trastuzumab, and some other drugs [86-88]. There are also targeted drugs inhibit angiogenesis directly such as endostatin and angiostatin, the two endogenous tumor neovascular inhibitors that inhibit tumor angiogenesis, induce tumor cell apoptosis, prevent tumor invasion and metastasis by inhibiting the growth of tumor endothelial cells [89]. Both drugs inhibit angiogenesis directly, being used in clinical practice in China [90].

Sorafenib is a multi-target inhibitor inhibit targets for Fms-like tyrosine kinase, c-KIT, platelet-derived growth factor receptor, and Raf/MEK/ERK signaling pathways [91-93]. Acting on multiple targets can not only inhibit tumor cell growth and differentiation but also inhibit tumor neovascularization [94] and improve treatment efficiency, so it can be used for the treatment of NSCLC and liver cancer [95].

Mammalian target of rapamycin (mTOR) inhibitors have been used as immunosuppressants for more than 10 years clinically [96], which is a serine/threonine protein kinase involved in regulating cell proliferation, growth, and metabolism [97,98].

Other targeted antitumor drugs such as histone deacetylase inhibitors mainly control gene expression by changing the histone acetylation degree, then changing the chromatin structure [99], which could induce tumor cell growth arrest, differentiation, and apoptosis to treattumors [100]. For example, the vorinostat of Merckwas the first histone deacetylase inhibitor approved by the US Food and Drug Administration in treating cutaneous T-cell lymphoma, marking a new approach to tumor therapy [101].

Autophagy is a catabolic process that leads to cellular degradation and the recycling of proteins and organelles by lysosomal digestion. Induced in starvation and several forms of stress rapidly, in which its dysregulation would join the processes like tumor. However, Apoptosis is a type of programmed cell death that cells are destroyed without releasing noxious substances into the surrounding area. Although the molecular mechanisms of apoptosis still need clarification, several proteins have been already known to have a vital role in regulating programmed cell death. It was proposed that therapeutic resistance in cancer is due to an upregulation of anti-apoptotic proteins and a downregulation of pro-apoptotic proteins, leading to genetic instability and the activation of oncogenes that favor cell survival and resistance to chemotherapy and recurrence (Figure 4).

Figure 4.

Figure 4

Status of proteins that participate in the apoptotic pathway in cancer. An overexpression of anti-apoptotic proteins has been reported, as well as a downregulation of pro-apoptotic proteins that participate in the mitochondrial apoptotic pathway and in the TNF receptor pathway. It has been suggested that the dysregulation of these proteins induces resistance to apoptosis in different therapeutic approaches.

Genetic engineering

At the end of the 20th century, a series of significant discoveries in cell and molecular biology promoted the biomedical technology development and many technological breakthroughs [102] including the found of wild-type tumor suppressor genes, suicide genes, anti-drug resistance genes and antisense oligonucleotides, and tumor genetically engineered bacteria tumors [103-105]. For example, Herceptin is a recombinant DNA-derived humanized monoclonal chimeric anti-p185HER-2 antibody [106] that can specifically bind to HER-2, downregulate it’s gene, antagonize the growth-promoting effects of it’s family, and mediate antibody-dependent cytotoxicity and anti-angiogenesis [107]. G3139 is an antisense oligonucleotide drug against Bcl-2 [108,109]. The renewal of these technologies have led to radical treatments for cancer (Figure 5).

Figure 5.

Figure 5

Comparison of sources and technologies for anti-cancer compounds. Traditional sources and new technologies that use microbial fermentation, insect and mammalian cell cultures, and transgenic animals have drawbacks in cost, scalability, product safety, and authenticity.

In recent years, the development of molecular biology technology has been rapidly changed [110]. New cancer vaccine have become the focus, including genetically engineered cancer vaccines. Studies have shown that genetically modified tumor cells can kill cancer cells efficiently [111]. At present, the clinical application of cancer vaccines mainly focuses on the treatment of lung cancer, malignant melanoma, colon cancer, and certain hematological tumors [112,113]. Therefore, the use of appropriate anticancer drugs for combination and sequential chemotherapy to achieve the goal of curing cancer or prolonging life is an optimized protocol.

Nanotechnology

Nanoparticles refer to particles between 1 and 100 nm in size, which havestrong adsorption capacity, large surface area, high catalytic efficiency, and high surface reactivity [114]. It is a new way to deliver anticancer drugs. Particles >200 nm in tissue are easily phagocytized by the phagocytic system, whereas magnetic particles <100 nm are more easily adsorbed and deposited at a lower rate, facilitating diffusion to tissues are widely used in antitumor drugs [115]. The properties exhibited by nanomaterials indicate anopolymer material showed broad anticancer application.

Other ways

With the further research of physiological and biochemical mechanisms, some drugs known as prototype drugs have achieved great success in medical effects and in the pharmaceutical market [117]. Many drugs with intellectual property rights have emerged, and those drugs with the same efficacy are called “me-too” drugs [118], the research of it is to find similar chemical structures that are not protected by patents [119]. Researchers change local chemical structures, increase water solubility or fat solubility, and bioavailability [120]. Those drugs would cause metabolic transformation in vivo, prolonging the duration of action, which are sometimes better than the original drug, reducing side effects and adverse reactions to some extent [121]. For example, Melphalan (sarcolysin), with phenylalanine as the carrier, has a better effect on malignant tumors [122]. Formylmerphalan is obtained by subjecting NH2 to formylation on the basis of melphalan [123], comparing with sarcolysine, it has higher therapeutic index and lower toxicity [124]. These drugs have followed the development ideas, mechanism of action, and targets of innovative drugs [125], and have modified the listed drugs in chemical structure, circumventing patent infringement with low research difficulty, low investment, and low risk. It is a way researching on new drugs, and it is also a shortcut to create a transition [126].

Old drugs refer to drugs that have been put on the market for clinical application and are known to everyone, Basedon previous research and development, detailed molecular structure, mechanism of action, and safety information [127]. “Me-too” dugs means shortening the development cycle of small molecule drugs, reducing risks, and increasing the success rate of small molecule drug development, enabling faster entry into clinical trials and rapid phase II clinical trials [128]. The assessment is reported to save approximately 40% of the cost and shorten the development cycle to 3 to 12 years [129,130].

Challenges and opportunities

Since the 20th century, people havn’t paid much attention to oncology drug research. Traditional cytotoxic drugs are still the main body of cancer drug therapy. However, with the development of molecular oncology and molecular pharmacology, the nature of tumor is gradually being clarified. The application of advanced technologies such as large-scale rapid screening, combinatorial chemistry, and genetic engineering have accelerated the drug development process [131,132].

However, at present, the development of small molecule drugs has reached a bottle neck, which is not only reflected in the floating number of new drugs on the market, but also in the increasing number of pharmaceutical R & D enterprises invest without breakthrough. For small molecule drugs, it is a matter of time to catch up or surpass. In the next few decades, the market share of large molecule drugs will be higher, gradually surpassing small molecule drugsbut does not mean that small molecule drugs will disappear. It just means that such drugs will start to move forward steadily rather than speedily [133-135].

Conclusion

Although therapeutic drugs have been discovered by this traditional method, there are still problems such as unpredictability, blindness, and resource inefficiency. When life sciences enter the post-genome era, scientists will discover new genes from a large number of gene sequencing results, delve into their functions and regulatory networks, andimprove the quality and efficiency of innovative drug research through a large number of bioinformatics libraries, compound information libraries and biochips.

Acknowledgements

This study was supported partly by grants from the National Natural Science Foundation of China (81772932, 81972214, 81472202 and 81302065), Shanghai Natural Science Foundation (20ZR1472400), Natural Science Foundation of Hunan Province of China (2018JJ4061, 2020JJ4278 and 2020JJ5127), the Key Program of Hunan Provincial Department of Science and Technology (2020WK2020 and 2019NK2111), the Scientific Research Fund of Hunan Provincial Education Department (18B290), Zhuzhou Key Science & Technology Program of Hunan Province (2017 and 2019), Construction of Clinical Medical Centre for Tumor Biological Samples in Nantong (HS2016004) and Jiangsu 333 Program (BRA2017205).

Disclosure of conflict of interest

None.

References

  • 1.Wang L, Qin W, Huo YJ, Li X, Shi Q, Rasko JEJ, Janin A, Zhao WL. Advances in targeted therapy for malignant lymphoma. Signal Transduct Target Ther. 2020;5:15. doi: 10.1038/s41392-020-0113-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Patel J, Baptiste BA, Kim E, Hussain M, Croteau DL, Bohr VA. DNA damage and mitochondria in cancer and aging. Carcinogenesis. 2020;41:1625–1634. doi: 10.1093/carcin/bgaa114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Cristóbal I, Sanz-Alvarez M, Torrejón B, Santos A, Luque M, Rojo F, García-Foncillas J. Potential therapeutic impact of mir-145 deregulation in colorectal cancer. Mol Ther. 2018;26:1399–1400. doi: 10.1016/j.ymthe.2018.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Li XN, Wang ZJ, Ye CX, Zhao BC, Li ZL, Yang Y. RNA sequencing reveals the expression profiles of circRNA and indicates that circDDX17 acts as a tumor suppressor in colorectal cancer. J Exp Clin Cancer Res. 2018;37:325. doi: 10.1186/s13046-018-1006-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Su R, Cao S, Ma J, Liu Y, Liu X, Zheng J, Chen J, Liu L, Cai H, Li Z, Zhao L, He Q, Xue Y. Knockdown of SOX2OT inhibits the malignant biological behaviors of glioblastoma stem cells via up-regulating the expression of miR-194-5p and miR-12. Mol Cancer. 2018;16:171. doi: 10.1186/s12943-017-0737-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Zhu Y, Wang C, Becker SA, Hurst K, Nogueira LM, Findlay VJ, Camp ER. miR-145 antagonizes SNAI1-mediated stemness and radiation resistance in colorectal cancer. Mol Ther. 2018;26:744–754. doi: 10.1016/j.ymthe.2017.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bhatta B, Cooks T. Reshaping the tumor microenvironment: extracellular vesicles as messengers of cancer cells. Carcinogenesis. 2020;41:1461–1470. doi: 10.1093/carcin/bgaa107. [DOI] [PubMed] [Google Scholar]
  • 8.Du H, Jia A, Gu M, Li S, Zhang P, Bai J. Thyrotoxicosis and osteomalacia: from symptom to pathogenesis. Am J Transl Res. 2021;13:803–811. [PMC free article] [PubMed] [Google Scholar]
  • 9.Wang M, Yu F, Chen X, Li P, Wang K. The underlying mechanisms of noncoding RNAs in the chemoresistance of hepatocellular carcinoma. Mol Ther Nucleic Acids. 2020;21:13–27. doi: 10.1016/j.omtn.2020.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Elsedawy NB, Nace RA, Russell SJ, Schulze AJ. Oncolytic activity of targeted picornaviruses formulated as synthetic infectious RNA. Mol Ther Oncolytics. 2020;17:484–495. doi: 10.1016/j.omto.2020.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zhu Y, Wang C, Becker SA, Hurst K, Nogueira LM, Findlay VJ, Camp ER. miR-145 antagonizes SNAI1-mediated stemness and radiation resistance in colorectal cancer. Mol Ther. 2018;26:744–754. doi: 10.1016/j.ymthe.2017.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chen W, Zheng R, Zhang S, Zeng H, Xia C, Zuo T, Yang Z, Zou X, He J. Cancer incidence and mortality in China, 2013. Cancer Lett. 2017;401:63–71. doi: 10.1016/j.canlet.2017.04.024. [DOI] [PubMed] [Google Scholar]
  • 13.Samet JM, Niwa O. At the 75th anniversary of the bombings of Hiroshima and Nagasaki, the Radiation Effects Research Foundation continues studies of the atomic bomb survivors and their children. Carcinogenesis. 2020;41:1471–1472. doi: 10.1093/carcin/bgaa104. [DOI] [PubMed] [Google Scholar]
  • 14.Sun Q, Wang W. Structures of BRAF-MEK1-14-3-3 sheds light on drug discovery. Signal Transduct Target Ther. 2019;4:59. doi: 10.1038/s41392-019-0096-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ma YS, Huang T, Zhong XM, Zhang HW, Cong XL, Xu H, Lu GX, Yu F, Xue SB, Lv ZW, Fu D. Proteogenomic characterization and comprehensive integrative genomic analysis of human colorectal cancer liver metastasis. Mol Cancer. 2018;17:139. doi: 10.1186/s12943-018-0890-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Oshima G, Guo N, He C, Stack ME, Poon C, Uppal A, Wightman SC. In vivo delivery and therapeutic effects of a MicroRNA on colorectal liver metastases. Mol Ther. 2018;25:1588–1595. doi: 10.1016/j.ymthe.2017.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gao T, Hu Q, Hu X, Lei Q, Feng Z, Yu X. Novel selective TOPK inhibitor SKLB-C05 inhibits colorectal carcinoma growth and metastasis. Cancer Lett. 2019;445:11–23. doi: 10.1016/j.canlet.2018.12.016. [DOI] [PubMed] [Google Scholar]
  • 18.Terasaki M, Uehara O, Ogasa S, Sano T, Kubota A, Kojima H, Tanaka T, Maeda H, Miyashita K, Mutoh M. Alteration of fecal microbiota by fucoxanthin results in prevention of colorectal cancer in AOM/DSS mice. Carcinogenesis. 2021;42:210–219. doi: 10.1093/carcin/bgaa100. [DOI] [PubMed] [Google Scholar]
  • 19.Murillo-Garzón V, Gorroño-Etxebarria I, Åkerfelt M, Puustinen MC, Sistonen L, Nees M, Carton J, Waxman J, Kypta RM. Frizzled-8 integrates Wnt-11 and transforming growth factor-β signaling in prostate cancer. Nat Commun. 2018;9:1747. doi: 10.1038/s41467-018-04042-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bao Z, Zheng Q, Li L. Oncogenic roles and mechanisms of lncRNA AGAP2-AS1 in human solid tumors. Am J Transl Res. 2021;13:757–769. [PMC free article] [PubMed] [Google Scholar]
  • 21.He Y, Chen D, Yi Y, Zeng S, Liu S, Li P, Xie H, Yu P, Jiang G, Liu H. Histone deacetylase inhibitor sensitizes ERCC1-high non-small-cell lung cancer cells to cisplatin via regulating miR-149. Mol Ther Oncolytics. 2020;17:448–459. doi: 10.1016/j.omto.2020.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Wu X, Pan Y, Fang Y, Zhang J, Xie M, Yang F, Yu T, Ma P, Li W, Shu Y. The biogenesis and functions of piRNAs in human diseases. Mol Ther Nucleic Acids. 2020;21:108–120. doi: 10.1016/j.omtn.2020.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Wei F, Zhang T, Deng SC, Wei JC, Yang P, Wang Q, Chen ZP, Li WL, Chen HC, Hu H, Cao J. PD-L1 promotes colorectal cancer stem cell expansion by activating HMGA1-dependent signaling pathways. Cancer Lett. 2019;S0304-3835:30100–30109. doi: 10.1016/j.canlet.2019.02.022. [DOI] [PubMed] [Google Scholar]
  • 24.Hu Z, Zheng X, Jiao D, Zhou Y, Sun R, Wang B, Tian Z, Wei H. LunX-CAR T cells as a targeted therapy for non-small cell lung cancer. Mol Ther Oncolytics. 2020;17:361–370. doi: 10.1016/j.omto.2020.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sadremomtaz A, Ali AM, Jouyandeh F, Balalaie S, Navari R, Broussy S, Mansouri K, Groves MR, Asghari SM. Molecular docking, synthesis and biological evaluation of Vascular Endothelial Growth Factor (VEGF) B based peptide as antiangiogenic agent targeting the second domain of the Vascular Endothelial Growth Factor Receptor 1 (VEGFR1D2) for anticancer application. Signal Transduct Target Ther. 2020;5:76. doi: 10.1038/s41392-020-0177-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Rezvani K, Rouce R, Liu E, Shpall E. Engineering natural killer cells for cancer immunotherapy. Mol Ther. 2017;25:1769–1781. doi: 10.1016/j.ymthe.2017.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cullis PR, Hope MJ. Lipid nanoparticle systems for enabling gene therapies. Mol Ther. 2017;25:1467–1475. doi: 10.1016/j.ymthe.2017.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Li Y, Zhang X, Zheng Q, Zhang Y, Ma Y, Zhu C, Yang L, Peng X, Wang Q, Wang B, Meng X, Li H, Liu J. YAP1 inhibition in HUVECs is associated with released exosomes and increased hepatocarcinoma invasion and metastasis. Mol Ther Nucleic Acids. 2020;21:86–97. doi: 10.1016/j.omtn.2020.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 29.You JH, Lee J, Roh JL. Mitochondrial pyruvate carrier 1 regulates ferroptosis in drug-tolerant persister head and neck cancer cells via epithelial-mesenchymal transition. Cancer Lett. 2021;507:40–54. doi: 10.1016/j.canlet.2021.03.013. [DOI] [PubMed] [Google Scholar]
  • 30.Ma YS, Lv ZW, Yu F, Chang ZY, Cong XL, Zhong XM, Lu GX, Zhu J, Fu D. MiRNA-302a/d inhibits the self-renewal capability and cell cycle entry of liver cancer stem cells by targeting the E2F7/AKT axis. J Exp Clin Cancer Res. 2018;37:252. doi: 10.1186/s13046-018-0927-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang Z, Qin J, Zhao J, Li J, Li D, Popp M, Popp F, Alakus H, Kong B, Dong Q, Nelson PJ, Zhao Y, Bruns CJ. Inflammatory IFIT3 renders chemotherapy resistance by regulating post-translational modification of VDAC2 in pancreatic cancer. Theranostics. 2020;10:7178–7192. doi: 10.7150/thno.43093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ma J, Kala S, Yung S, Chan TM, Cao Y, Jiang Y, Liu X, Giorgio S, Peng L, Wong AST. Blocking stemness and metastatic properties of ovarian cancer cells by targeting p70S6K with dendrimer nanovector-based siRNA delivery. Mol Ther. 2018;26:70–83. doi: 10.1016/j.ymthe.2017.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Deng S, Deng Q, Zhang Y, Ye H, Yu X, Zhang Y, Han GY, Luo P, Wu M, Yu Y, Han W. Non-platelet-derived CXCL4 differentially regulates cytotoxic and regulatory T cells through CXCR3 to suppress the immune response to colon cancer. Cancer Lett. 2019;443:1–12. doi: 10.1016/j.canlet.2018.11.017. [DOI] [PubMed] [Google Scholar]
  • 34.Di L, Liu LJ, Yan YM, Fu R, Li Y, Xu Y, Cheng YX, Wu ZQ. Discovery of a natural small-molecule compound that suppresses tumor EMT, stemness and metastasis by inhibiting TGFβ/BMP signaling in triple-negative breast cancer. J Exp Clin Cancer Res. 2019;38:134. doi: 10.1186/s13046-019-1130-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Yu F, Liu JB, Wu ZJ, Xie WT, Zhong XJ, Hou LK, Wu W, Lu HM, Jiang XH, Jiang JJ, Cao ZY, Cong GJ, Shi MX, Jia CY, Lu GX, Song YC, Chai L, Lv ZW, Wu CY, Ma YS, Fu D. Tumor suppressive microRNA-124a inhibits stemness and enhances gefitinib sensitivity of non-small cell lung cancer cells by targeting ubiquitin-specific protease 1. Cancer Lett. 2018;427:74–84. doi: 10.1016/j.canlet.2018.04.022. [DOI] [PubMed] [Google Scholar]
  • 36.Rizzo M. Mechanisms of docetaxel resistance in prostate cancer: the key role played by miRNAs. Biochim Biophys Acta Rev Cancer. 2021;1875:188481. doi: 10.1016/j.bbcan.2020.188481. [DOI] [PubMed] [Google Scholar]
  • 37.Wang X, Yu H, Sun W, Kong J, Zhang L, Tang J, Wang J, Xu E, Lai M, Zhang H. The long non-coding RNA CYTOR drives colorectal cancer progression by interacting with NCL and Sam68. Mol Cancer. 2018;17:110. doi: 10.1186/s12943-018-0860-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wang Y, Wu M, Lei Z, Huang M, Li Z, Wang L. Dysregulation of miR-6868-5p/FOXM1 circuit contributes to colorectal cancer angiogenesis. J Exp Clin Cancer Res. 2018;37:292. doi: 10.1186/s13046-018-0970-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chen L, Quan H, Xu Z, Wang H, Xia Y, Lou L, Yang W. A modular biomimetic strategy for the synthesis of macrolide P-glycoprotein inhibitors via Rh-catalyzed C-H activation. Nat Commun. 2020;11:2151. doi: 10.1038/s41467-020-16084-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lin J, Liao S, Li E, Liu Z, Zheng R, Wu X, Zeng W. circCYFIP2 acts as a sponge of miR-1205 and affects the expression of its target gene E2F1 to regulate gastric cancer metastasis. Mol Ther Nucleic Acids. 2020;21:121–132. doi: 10.1016/j.omtn.2020.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Panda M, Tripathi SK, Biswal BK. SOX9: an emerging driving factor from cancer progression to drug resistance. Biochim Biophys Acta Rev Cancer. 2021;1875:188517. doi: 10.1016/j.bbcan.2021.188517. [DOI] [PubMed] [Google Scholar]
  • 42.Dong R, Gong Y, Meng W, Yuan M, Zhu H, Ying M, He Q, Cao J, Yang B. The involvement of M2 macrophage polarization inhibition in fenretinide-mediated chemopreventive effects on colon cancer. Cancer Lett. 2017;388:43–53. doi: 10.1016/j.canlet.2016.11.029. [DOI] [PubMed] [Google Scholar]
  • 43.Wang J, Yang S, Ji Q, Li Q, Zhou F, Li Y, Yuan F, Liu J, Tian Y, Zhao Y, Zheng Y. Long non-coding RNA EPIC1 promotes cell proliferation and motility and drug resistance in glioma. Mol Ther Oncolytics. 2020;17:130–137. doi: 10.1016/j.omto.2020.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ylä-Herttuala S. The pharmacology of gene therapy. Mol Ther. 2017;25:1731–1732. doi: 10.1016/j.ymthe.2017.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Park GB, Jeong JY, Kim D. GLUT5 regulation by AKT1/3-miR-125b-5p downregulation induces migratory activity and drug resistance in TLR-modified colorectal cancer cells. Carcinogenesis. 2020;41:1329–1340. doi: 10.1093/carcin/bgaa074. [DOI] [PubMed] [Google Scholar]
  • 46.Huang X, She L, Liu H, Liu P, Chen J, Chen Y, Zhou W, Lu Y, Lin J. Study of oral microorganisms contributing to non-carious cervical lesions via bacterial interaction and pH regulation. J Cell Mol Med. 2021;25:3103–3112. doi: 10.1111/jcmm.16370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Holmes TR, Al Matouq J, Holmes M, Sioda N, Rudd JC, Bloom C, Nicola L, Palermo NY, Madson JG, Lovas S, Hansen LA. Targeting 14-3-3ε activates apoptotic signaling to prevent cutaneous squamous cell carcinoma. Carcinogenesis. 2021;42:232–242. doi: 10.1093/carcin/bgaa091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Shan G, Shao B, Liu Q, Zeng Y, Fu C, Chen A, Chen Q. circFMN2 sponges miR-1238 to promote the expression of LIM-homeobox gene 2 in prostate cancer cells. Mol Ther Nucleic Acids. 2020;21:133–146. doi: 10.1016/j.omtn.2020.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 49.Wang D, Hu G, Du Y, Zhang C, Lu Q, Lv N, Luo S. Aberrant activation of hedgehog signaling promotes cell proliferation via the transcriptional activation of forkhead Box M1 in colorectal cancer cells. J Exp Clin Cancer Res. 2017;36:23. doi: 10.1186/s13046-017-0491-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Li T, Jian X, He H, Lai Q, Li X, Deng D, Liu T, Zhu J, Jiao H, Ye Y, Wang S, Yang M, Zheng L, Zhou W, Ding Y. MiR-452 promotes an aggressive colorectal cancer phenotype by regulating a Wnt/β-catenin positive feedback loop. J Exp Clin Cancer Res. 2018;37:238. doi: 10.1186/s13046-018-0879-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ma YS, Yu F, Zhong XM, Lu GX, Cong XL, Xue SB, Xie WT, Hou LK, Pang LJ, Wu W, Zhang W, Cong LL, Liu T, Long HD, Sun R, Sun HY, Lv ZW, Wu CY, Fu D. miR-30 family reduction maintains self-renewal and promotes tumorigenesis in NSCLC-initiating cells by targeting oncogene TM4SF. Mol Ther. 2018;26:30447–7. doi: 10.1016/j.ymthe.2018.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Tsai KW, Lo YH, Liu H, Yeh CY, Chen YZ, Hsu CW, Chen WS, Wang JH. Linc00659, a long noncoding RNA, acts as novel oncogene in regulating cancer cell growth in colorectal cancer. Mol Cancer. 2018;17:72. doi: 10.1186/s12943-018-0821-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Duan S, Huang W, Liu X, Liu X, Chen N, Xu Q, Hu Y, Song W, Zhou J. IMPDH2 promotes colorectal cancer progression through activation of the PI3K/AKT/mTOR and PI3K/AKT/FOXO1 signaling pathways. J Exp Clin Cancer Res. 2018;37:304. doi: 10.1186/s13046-018-0980-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jeong S, Yun HK, Jeong YA, Jo MJ, Kang SH, Kim JL, Kim DY, Park SH, Kim BR, Na YJ, Lee SI, Kim HD, Kim DH, Oh SC, Lee DH. Cannabidiol-induced apoptosis is mediated by activation of Noxa in human colorectal cancer cells. Cancer Lett. 2019;447:12–23. doi: 10.1016/j.canlet.2019.01.011. [DOI] [PubMed] [Google Scholar]
  • 55.Li T, Jian X, He H, Lai Q, Li X, Deng D. MiR-452 promotes an aggressive colorectal cancer phenotype by regulating a Wnt/β-catenin positive feedback loop. J Exp Clin Cancer Res. 2018;37:238. doi: 10.1186/s13046-018-0879-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Yuki R, Hagino M, Ueno S, Kuga T, Saito Y, Fukumoto Y, Yamaguchi N, Yamaguchi N, Nakayama Y. The tyrosine kinase v-Src modifies cytotoxicities of anticancer drugs targeting cell division. J Cell Mol Med. 2021;25:1677–1687. doi: 10.1111/jcmm.16270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.de Bruijn HS, Mashayekhi V, Schreurs TJL, van Driel PBAA, Strijkers GJ, van Diest PJ, Lowik CWGM, Seynhaeve ALB, Hagen TLMT, Prompers JJ, Henegouwen PMPVBE, Robinson DJ, Oliveira S. Acute cellular and vascular responses to photodynamic therapy using EGFR-targeted nanobody-photosensitizer conjugates studied with intravital optical imaging and magnetic resonance imaging. Theranostics. 2020;10:2436–2452. doi: 10.7150/thno.37949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ratnayake G, Bain AL, Fletcher N, Howard CB, Khanna KK, Thurecht KJ. RNA interference to enhance radiation therapy: targeting the DNA damage response. Cancer Lett. 2018;439:14–23. doi: 10.1016/j.canlet.2018.09.011. [DOI] [PubMed] [Google Scholar]
  • 59.Piramoon M, Khodadust F, Hosseinimehr SJ. Radiolabeled nanobodies for tumor targeting: from bioengineering to imaging and therapy. Biochim Biophys Acta Rev Cancer. 2021;1875:188529. doi: 10.1016/j.bbcan.2021.188529. [DOI] [PubMed] [Google Scholar]
  • 60.Zhang J, Ren P, Xu D, Liu X, Liu Z, Zhang C, Li Y, Wang L, Du X, Xing B. Human UTP14a promotes colorectal cancer progression by forming a positive regulation loop with c-Myc. Cancer Lett. 2019;440-441:106–115. doi: 10.1016/j.canlet.2018.10.010. [DOI] [PubMed] [Google Scholar]
  • 61.Giordano G, Parcesepe P, D’Andrea MR, Coppola L, Di Raimo T, Remo A, Manfrin E, Fiorini C, Scarpa A, Amoreo CA, Conciatori F, Milella M, Caruso FP, Cerulo L, Porras A, Pancione M. JAK/Stat5-mediated subtype-specific lymphocyte antigen 6 complex, locus G6D (LY6G6D) expression drives mismatch repair proficient colorectal cancer. J Exp Clin Cancer Res. 2019;38:28. doi: 10.1186/s13046-018-1019-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ke W, Saba JA, Yao CH, Hilzendeger MA, Drangowska-Way A, Joshi C, Mony VK, Benjamin SB, Zhang S, Locasale J, Patti GJ, Lewis N, O’Rourke EJ. Dietary serine-microbiota interaction enhances chemotherapeutic toxicity without altering drug conversion. Nat Commun. 2020;11:2587. doi: 10.1038/s41467-020-16220-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Peng Y, Kajiyama H, Yuan H, Nakamura K, Yoshihara M, Yokoi A, Fujikake K, Yasui H, Yoshikawa N, Suzuki S, Senga T, Shibata K, Kikkawa F. PAI-1 secreted from metastatic ovarian cancer cells triggers the tumor-promoting role of the mesothelium in a feedback loop to accelerate peritoneal dissemination. Cancer Lett. 2019;442:181–192. doi: 10.1016/j.canlet.2018.10.027. [DOI] [PubMed] [Google Scholar]
  • 64.Chen F, Wu X, Niculite C, Gilca M, Petrusca D, Rogozea A, Rice S, Guo B, Griffin S, Calin GA, Boswell HS, Konig H. Classic and targeted anti-leukaemic agents interfere with the cholesterol biogenesis metagene in acute myeloid leukaemia: therapeutic implications. J Cell Mol Med. 2020;24:7378–92. doi: 10.1111/jcmm.15339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Wei F, Zhang T, Deng SC, Wei JC, Yang P, Wang Q, Chen ZP, Li WL, Chen HC, Hu H, Cao J. PD-L1 promotes colorectal cancer stem cell expansion by activating HMGA1-dependent signaling pathways. Cancer Lett. 2019;S0304-3835:30100–4. doi: 10.1016/j.canlet.2019.02.022. [DOI] [PubMed] [Google Scholar]
  • 66.Parry AJ, Hoare M, Bihary D, Hänsel-Hertsch R, Smith S, Tomimatsu K, Mannion E, Smith A, D’Santos P, Russell IA, Balasubramanian S, Kimura H, Samarajiwa SA, Narita M. NOTCH-mediated non-cell autonomous regulation of chromatin structure during senescence. Nat Commun. 2018;9:1840. doi: 10.1038/s41467-018-04283-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Al-Bari MAA. Co-targeting of lysosome and mitophagy in cancer stem cells with chloroquine analogues and antibiotics. J Cell Mol Med. 2020;24:11667–11679. doi: 10.1111/jcmm.15879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Luo Y, Chen JJ, Lv Q, Qin J, Huang YZ, Yu MH, Zhong M. Long non-coding RNA NEAT1 promotes colorectal cancer progression by competitively binding miR-34a with SIRT1 and enhancing the Wnt/β-catenin signaling pathway. Cancer Lett. 2019;440-441:11–22. doi: 10.1016/j.canlet.2018.10.002. [DOI] [PubMed] [Google Scholar]
  • 69.De Robertis M, Mazza T, Fusilli C, Loiacono L, Poeta ML, Sanchez M, Massi E, Lamorte G, Diodoro MG, Pescarmona E, Signori E, Pesole G, Vescovi AL, Garcia-Foncillas J, Fazio VM. EphB2 stem-related and EphA2 progression-related miRNA-based networks in progressive stages of CRC evolution: clinical significance and potential miRNA drivers. Mol Cancer. 2018;17:169. doi: 10.1186/s12943-018-0912-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Bouclier C, Simon M, Laconde G, Pellerano M, Diot S, Lantuejoul S, Busser B, Vanwonterghem L, Vollaire J, Josserand V, Legrand B, Coll JL, Amblard M, Hurbin A, Morris MC. Stapled peptide targeting the CDK4/Cyclin D interface combined with Abemaciclib inhibits KRAS mutant lung cancer growth. Theranostics. 2020;10:2008–2028. doi: 10.7150/thno.40971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Xing L, Mi W, Zhang Y, Tian S, Zhang Y, Qi R, Lou G, Zhang C. The identification of six risk genes for ovarian cancer platinum response based on global network algorithm and verification analysis. J Cell Mol Med. 2020;24:9839–9852. doi: 10.1111/jcmm.15567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bae SDW, Nguyen R, Qiao L, George J. Role of the constitutive androstane receptor (CAR) in human liver cancer. Biochim Biophys Acta Rev Cancer. 2021;1875:188516. doi: 10.1016/j.bbcan.2021.188516. [DOI] [PubMed] [Google Scholar]
  • 73.Belhadj S, Moutinho C, Mur P, Setien F, Llinàs-Arias P, Pérez-Salvia M. Germline variation in O6-methylguanine-DNA methyltransferase (MGMT) as cause of hereditary colorectal cancer. Cancer Lett. 2019;447:86–92. doi: 10.1016/j.canlet.2019.01.019. [DOI] [PubMed] [Google Scholar]
  • 74.Tsai YC, Zeng T, Abou-Kheir W, Yeh HL, Yin JJ, Lee YC, Chen WY, Liu YN. Disruption of ETV6 leads to TWIST1-dependent progression and resistance to epidermal growth factor receptor tyrosine kinase inhibitors in prostate cancer. Mol Cancer. 2018;17:42. doi: 10.1186/s12943-018-0785-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Vitiello PP, Cardone C, Martini G, Ciardiello D, Belli V, Matrone N, Barra G. Receptor tyrosine kinase-dependent PI3K activation is an escape mechanism to vertical suppression of the EGFR/RAS/MAPK pathway in KRAS-mutated human colorectal cancer cell lines. J Exp Clin Cancer Res. 2019;38:41. doi: 10.1186/s13046-019-1035-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Sun J, Luo Q, Liu L, Song G. Low-level shear stress promotes migration of liver cancer stem cells via the FAK-ERK1/2 signalling pathway. Cancer Lett. 2018;427:1–8. doi: 10.1016/j.canlet.2018.04.015. [DOI] [PubMed] [Google Scholar]
  • 77.Yu J, Han Z, Sun Z, Wang Y, Zheng M, Song C. LncRNA SLCO4A1-AS1 facilitates growth and metastasis of colorectal cancer through β-catenin-dependent Wnt pathway. J Exp Clin Cancer Res. 2018;37:222. doi: 10.1186/s13046-018-0896-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Fassunke J, Müller F, Keul M, Michels S, Dammert MA, Schmitt A. Overcoming EGFRG724S-mediated osimertinib resistance through unique binding characteristics of second-generation EGFR inhibitors. Nat Commun. 2018;9:4655. doi: 10.1038/s41467-018-07078-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Tzchori I, Falah M, Shteynberg D, Levin Ashkenazi D, Loberman Z, Perry L, Flugelman MY. Improved patency of ePTFE grafts as a hemodialysis access site by seeding autologous endothelial cells expressing fibulin-5 and VEGF. Mol Ther. 2018;26:1660–1668. doi: 10.1016/j.ymthe.2018.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Yue B, Liu C, Sun H, Liu M, Song C, Cui R, Qiu S, Zhong M. A positive feed-forward loop between LncRNA-CYTOR and Wnt/β-catenin signaling promotes metastasis of colon cancer. Mol Ther. 2018;26:1287–1298. doi: 10.1016/j.ymthe.2018.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wang J, Yang S, Ji Q, Li Q, Zhou F, Li Y, Yuan F, Liu J, Tian Y, Zhao Y, Zheng Y. Long non-coding RNA EPIC1 promotes cell proliferation and motility and drug resistance in glioma. Mol Ther Oncolytics. 2020;17:130–137. doi: 10.1016/j.omto.2020.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Rauth S, Karmakar S, Batra SK, Ponnusamy MP. Recent advances in organoid development and applications in disease modeling. Biochim Biophys Acta Rev Cancer. 2021;1875:188527. doi: 10.1016/j.bbcan.2021.188527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Xu L, Huang J, Liu J, Xi Y, Zheng Z, To KKW, Chen Z, Wang F, Zhang Y, Fu L. CM082 enhances the efficacy of chemotherapeutic drugs by inhibiting the drug efflux function of ABCG2. Mol Ther Oncolytics. 2019;16:100–110. doi: 10.1016/j.omto.2019.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Jin F, Yang R, Wei Y, Wang D, Zhu Y, Wang X, Lu Y, Wang Y, Zen K, Li L. HIF-1α-induced miR-23a~27a~24 cluster promotes colorectal cancer progression via reprogramming metabolism. Cancer Lett. 2019;440-441:211–222. doi: 10.1016/j.canlet.2018.10.025. [DOI] [PubMed] [Google Scholar]
  • 85.Thomas D, Rathinavel AK, Radhakrishnan P. Altered glycosylation in cancer: a promising target for biomarkers and therapeutics. Biochim Biophys Acta Rev Cancer. 2021;1875:188464. doi: 10.1016/j.bbcan.2020.188464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Liu Y, Lv J, Liu J, Liang X, Jin X, Xie J. STAT3/p53 pathway activation disrupts IFN-β-induced dormancy in tumor-repopulating cells. J Clin Invest. 2018;128:1057–1073. doi: 10.1172/JCI96329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Zhang J, Xue W, Xu K, Yi L, Guo Y, Xie T, Tong H, Zhou B, Wang S, Li Q, Liu H, Chen X, Fang J, Zhang W. Dual inhibition of PFKFB3 and VEGF normalizes tumor vasculature, reduces lactate production, and improves chemotherapy in glioblastoma: insights from protein expression profiling and MRI. Theranostics. 2020;10:7245–7259. doi: 10.7150/thno.44427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Liu K, Wang X, Li D, Xu D, Li D, Lv Z, Zhao D, Chu WF, Wang XF. Ginkgolic acid, a SUMO-1 inhibitor, inhibits the progression of oral squamous cell carcinoma by alleviating SUMOylation of SMAD4. Mol Ther Oncolytics. 2019;16:86–99. doi: 10.1016/j.omto.2019.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Andrzejewska A, Dabrowska S, Nowak B, Walczak P, Lukomska B, Janowski M. Mesenchymal stem cells injected into carotid artery to target focal brain injury home to perivascular space. Theranostics. 2020;10:6615–6628. doi: 10.7150/thno.43169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zheng M, Huang J, Tong A, Yang H. Oncolytic viruses for cancer therapy: barriers and recent advances. Mol Ther Oncolytics. 2019;15:234–247. doi: 10.1016/j.omto.2019.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zhang S, Liu C, Zhang X. Mitochondrial damage mediated by miR-1 overexpression in cancer stem cells. Mol Ther Nucleic Acids. 2020;20:881. doi: 10.1016/j.omtn.2020.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Koujima T, Tazawa H, Ieda T, Araki H, Fushimi T, Shoji R, Kuroda S, Kikuchi S, Yoshida R, Umeda Y, Teraishi F, Urata Y, Mizuguchi H, Fujiwara T. Oncolytic virus-mediated targeting of the ERK signaling pathway inhibits invasive propensity in human pancreatic cancer. Mol Ther Oncolytics. 2020;17:107–117. doi: 10.1016/j.omto.2020.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Burton C, Bartee E. Syncytia formation in oncolytic virotherapy. Mol Ther Oncolytics. 2019;15:131–139. doi: 10.1016/j.omto.2019.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Weng H, Zeng L, Cao L, Chen T, Li Y, Xu Y, Peng Y, Ye Y. circFOXM1 contributes to sorafenib resistance of hepatocellular carcinoma cells by regulating MECP2 via miR-1324. Mol Ther Nucleic Acids. 2021;23:811–820. doi: 10.1016/j.omtn.2020.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Hu Y, Yu J, Wang Q, Zhang L, Chen X, Cao Y, Zhao J, Xu Y, Jiang D, Wang Y, Xiong W. Tartrate-resistant acid phosphatase 5/ACP5 interacts with p53 to control the expression of SMAD3 in lung adenocarcinoma. Mol Ther Oncolytics. 2020;16:272–288. doi: 10.1016/j.omto.2020.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Jin J, Zhao Q. Emerging role of mTOR in tumor immune contexture: impact on chemokine-related immune cells migration. Theranostics. 2020;10:6231–6244. doi: 10.7150/thno.45219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Chen Z, Li L, Wu W, Liu Z, Huang Y, Yang L, Luo Q, Chen J, Hou Y, Song G. Exercise protects proliferative muscle satellite cells against exhaustion via the Igfbp7-Akt-mTOR axis. Theranostics. 2020;10:6448–6466. doi: 10.7150/thno.43577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Fan H, Yu X, Zou Z, Zheng W, Deng X, Guo L, Jiang W, Zhan Q, Lu SH. Metformin suppresses the esophageal carcinogenesis in rats treated with NMBzA through inhibiting AMPK/mTOR signaling pathway. Carcinogenesis. 2019;40:669–679. doi: 10.1093/carcin/bgy160. [DOI] [PubMed] [Google Scholar]
  • 99.Han X, Yang J, Zeng F, Weng J, Zhang Y, Peng Q, Shen L, Ding S, Liu K, Gao Y. Programmable synthetic protein circuits for the identification and suppression of hepatocellular carcinoma. Mol Ther Oncolytics. 2020;17:70–82. doi: 10.1016/j.omto.2020.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Salem A, Alotaibi M, Mroueh R, Basheer HA, Afarinkia K. CCR7 as a therapeutic target in cancer. Biochim Biophys Acta Rev Cancer. 2021;1875:188499. doi: 10.1016/j.bbcan.2020.188499. [DOI] [PubMed] [Google Scholar]
  • 101.Hu F, Li H, Liu L, Xu F, Lai S, Luo X, Hu J, Yang X. Histone demethylase KDM4D promotes gastrointestinal stromal tumor progression through HIF1β/VEGFA signaling. Mol Cancer. 2018;17:107. doi: 10.1186/s12943-018-0861-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Herrera M, Llorens C, Rodríguez M, Herrera A, Ramos R, Gil B. Differential distribution and enrichment of non-coding RNAs in exosomes from normal and Cancer-associated fibroblasts in colorectal cancer. Mol Cancer. 2018;17:114. doi: 10.1186/s12943-018-0863-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Li J, Song P, Jiang T, Dai D, Wang H, Sun J, Zhu L, Xu W, Feng L, Shin VY, Morrison H, Wang X, Jin H. Heat shock factor 1 epigenetically stimulates glutaminase-1-dependent mTOR activation to promote colorectal carcinogenesis. Mol Ther. 2018;26:1828–1839. doi: 10.1016/j.ymthe.2018.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Li H, Zhou L, Zhou J, Li Q, Ji Q. Underlying mechanisms and drug intervention strategies for the tumour microenvironment. J Exp Clin Cancer Res. 2021;40:97. doi: 10.1186/s13046-021-01893-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Rodríguez MM, Fiore E, Bayo J, Atorrasagasti C, García M, Onorato A, Domínguez L, Malvicini M, Mazzolini G. Decreases CD47 expression on hepatic cancer stem cells and primes a potent antitumor T cell response induced by interleukin-12. Mol Ther. 2018;26:2738–2750. doi: 10.1016/j.ymthe.2018.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Tzelepis K, De Braekeleer E, Aspris D, Barbieri I, Vijayabaskar MS, Liu WH. SRPK1 maintains acute myeloid leukemia through effects on isoform usage of epigenetic regulators including BRD4. Nat Commun. 2018;9:5378. doi: 10.1038/s41467-018-07620-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Escobar G, Barbarossa L, Barbiera G, Norelli M, Genua M, Ranghetti A. Interferon gene therapy reprograms the leukemia microenvironment inducing protective immunity to multiple tumor antigens. Nat Commun. 2018;9:2896. doi: 10.1038/s41467-018-05315-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Shastri A, Choudhary G, Teixeira M, Gordon-Mitchell S, Ramachandra N, Bernard L. Antisense STAT3 inhibitor decreases viability of myelodysplastic and leukemic stem cells. J Clin Invest. 2018;128:5479–5488. doi: 10.1172/JCI120156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Yan C, Gu J, Jing H, Taishi J, Lee RJ. Tat-tagged and folate-modified N-succinyl-chitosan (Tat-Suc-FA) self-assembly nanoparticle for therapeutic delivery OGX-011 to A549 cells. Mol Pharm. 2017;14:1898–1905. doi: 10.1021/acs.molpharmaceut.6b01167. [DOI] [PubMed] [Google Scholar]
  • 110.Abou-El-Enein M, Grainger DW, Kili S. Registry contributions to strengthen cell and gene therapeutic evidence. Mol Ther. 2018;26:1172–1176. doi: 10.1016/j.ymthe.2018.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Petersen LM, Fenton JM, Kennedy LS, LaRochelle EPM, Bejarano S, Tsongalis GJ. HPV, vaccines, and cervical cancer in a low- and middle-income country. Curr Probl Cancer. 2020;44:100605. doi: 10.1016/j.currproblcancer.2020.100605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Deng S, Zhang G, Kuai J, Fan P, Wang X, Zhou P, Yang D, Zheng X, Liu X, Wu Q, Huang Y. Lentinan inhibits tumor angiogenesis via interferon γ and in a T cell independent manner. J Exp Clin Cancer Res. 2018;37:260. doi: 10.1186/s13046-018-0932-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Matsuyama T, Ishikawa T, Takahashi N, Yamada Y, Yasuno M, Kawano T, Uetake H, Goel A. Transcriptomic expression profiling identifies ITGBL, an epithelial to mesenchymal transition (EMT)-associated gene, is a promising recurrence prediction biomarker in colorectal cancer. Mol Cancer. 2019;18:19. doi: 10.1186/s12943-019-0945-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Toden S, Zumwalt TJ, Goel A. Non-coding RNAs and potential therapeutic targeting in cancer. Biochim Biophys Acta Rev Cancer. 2021;1875:188491. doi: 10.1016/j.bbcan.2020.188491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Luo Y, Qiao B, Zhang P, Yang C, Cao J, Yuan X, Ran H, Wang Z, Hao L, Cao Y, Ren J, Zhou Z. TME-activatable theranostic nanoplatform with ATP burning capability for tumor sensitization and synergistic therapy. Theranostics. 2020;10:6987–7001. doi: 10.7150/thno.44569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Wang YA, Li XL, Mo YZ, Fan CM, Tang L, Xiong F, Guo C, Xiang B, Zhou M, Ma J, Huang X, Wu X, Li Y, Li GY, Zeng ZY, Xiong W. Effects of tumor metabolic microenvironment on regulatory T cells. Mol Cancer. 2018;17:168. doi: 10.1186/s12943-018-0913-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Besnier M, Gasparino S, Vono R, Sangalli E, Facoetti A, Bollati V, Cantone L, Zaccagnini G, Maimone B, Fuschi P, Da Silva D, Schiavulli M, Aday S, Caputo M, Madeddu P, Emanueli C, Martelli F, Spinetti G. miR-210 enhances the therapeutic potential of bone-marrow-derived circulating proangiogenic cells in the setting of limb ischemia. Mol Ther. 2018;26:1694–1705. doi: 10.1016/j.ymthe.2018.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zuo D, Shogren KL, Zang J, Jewison DE, Waletzki BE, Miller AL 2nd, Okuno SH, Cai Z, Yaszemski MJ, Maran A. Inhibition of STAT3 blocks protein synthesis and tumor metastasis in osteosarcoma cells. J Exp Clin Cancer Res. 2018;37:244. doi: 10.1186/s13046-018-0914-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.González-Fernández Y, Brown HK, Patiño-García A, Heymann D, Blanco-Prieto MJ. Oral administration of edelfosine encapsulated lipid nanoparticles causes regression of lung metastases in pre-clinical models of osteosarcoma. Cancer Lett. 2018;430:193–200. doi: 10.1016/j.canlet.2018.05.030. [DOI] [PubMed] [Google Scholar]
  • 120.Gushchina LV, Bhattacharya S, McElhanon KE, Choi JH, Manring H, Beck EX, Alloush J, Weisleder N. Treatment with recombinant human MG53 protein increases membrane integrity in a mouse model of limb girdle muscular dystrophy 2B. Mol Ther. 2017;25:2360–2371. doi: 10.1016/j.ymthe.2017.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Yu TJ, Ma D, Liu YY, Xiao Y, Gong Y, Jiang YZ, Shao ZM, Hu X, Di GH. Bulk and single-cell transcriptome profiling reveal the metabolic heterogeneity in human breast cancers. Mol Ther. 2021 doi: 10.1016/j.ymthe.2021.03.003. S1525-0016(21)00132-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Martin-Tellez KS, van Houdt WJ, van Coevorden F, Colombo C, Fiore M. Isolated limb perfusion for soft tissue sarcoma: current practices and future directions. A survey of experts and a review of literature. Cancer Treat Rev. 2020;88:102058. doi: 10.1016/j.ctrv.2020.102058. [DOI] [PubMed] [Google Scholar]
  • 123.Lou X, Meng Y, Hou Y. A literature review on function and regulation mechanism of DKK4. J Cell Mol Med. 2021;25:2786–2794. doi: 10.1111/jcmm.16372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Oddone N, Boury F, Garcion E, Grabrucker AM, Martinez MC, Da Ros F, Janaszewska A, Forni F, Vandelli MA, Tosi G, Ruozi B, Duskey JT. Synthesis, characterization, and in vitro studies of an reactive oxygen species (ROS)-responsive methoxy polyethylene glycol-thioketal-melphalan prodrug for glioblastoma treatment. Front Pharmacol. 2020;11:574. doi: 10.3389/fphar.2020.00574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Murad JP, Tilakawardane D, Park AK, Lopez LS, Young CA, Gibson J, Yamaguchi Y, Lee HJ, Kennewick KT, Gittins BJ, Chang WC, Tran CP, Martinez C, Wu AM, Reiter RE, Dorff TB, Forman SJ, Priceman SJ. Pre-conditioning modifies the TME to enhance solid tumor CAR T cell efficacy and endogenous protective immunity. Mol Ther. 2021 doi: 10.1016/j.ymthe.2021.02.024. S1525-0016(21)00127-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Zhang F, Yan T, Guo W, Sun K, Wang S, Bao X, Liu K, Zheng B, Zhang H, Ren T. Novel oncogene COPS3 interacts with Beclin1 and Raf-1 to regulate metastasis of osteosarcoma through autophagy. J Exp Clin Cancer Res. 2018;37:135. doi: 10.1186/s13046-018-0791-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Wang X, Guo J, Yu P, Guo L, Mao X, Wang J, Miao S, Sun J. The roles of extracellular vesicles in the development, microenvironment, anticancer drug resistance, and therapy of head and neck squamous cell carcinoma. J Exp Clin Cancer Res. 2021;40:35. doi: 10.1186/s13046-021-01840-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Gambera S, Abarrategi A, González-Camacho F, Morales-Molina Á, Roma J, Alfranca A, García-Castro J. Clonal dynamics in osteosarcoma defined by RGB marking. Nat Commun. 2018;9:3994. doi: 10.1038/s41467-018-06401-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Terasaki M, Uehara O, Ogasa S, Sano T, Kubota A, Kojima H, Tanaka T, Maeda H, Miyashita K, Mutoh M. Alteration of fecal microbiota by fucoxanthin results in prevention of colorectal cancer in AOM/DSS mice. Carcinogenesis. 2021;42:210–219. doi: 10.1093/carcin/bgaa100. [DOI] [PubMed] [Google Scholar]
  • 130.Wang Y, Deng X, Yu C, Zhao G, Zhou J, Zhang G, Li M, Jiang D, Quan Z, Zhang Y. Synergistic inhibitory effects of capsaicin combined with cisplatin on human osteosarcoma in culture and in xenografts. J Exp Clin Cancer Res. 2018;37:251. doi: 10.1186/s13046-018-0922-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Wang LH, Tsai HC, Cheng YC, Lin CY, Huang YL, Tsai CH, Xu GH, Wang SW, Fong YC, Tang CH. CTGF promotes osteosarcoma angiogenesis by regulating miR-543/angiopoietin 2 signaling. Cancer Lett. 2017;391:28–37. doi: 10.1016/j.canlet.2017.01.013. [DOI] [PubMed] [Google Scholar]
  • 132.Aulova KS, Urusov AA, Sedykh SE, Toporkova LB, Lopatnikova JA, Buneva VN, Sennikov SV, Budde T, Meuth SG, Popova NA, Orlovskaya IA, Nevinsky GA. The association between EAE development in mice and the production of autoantibodies and abzymes after immunization of mice with different antigens. J Cell Mol Med. 2021;25:2493–2504. doi: 10.1111/jcmm.16183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Miao Y, Zheng Y, Geng Y, Yang L, Cao N, Dai Y, Wei Z. The role of GLS1-mediated glutaminolysis/2-HG/H3K4me3 and GSH/ROS signals in Th17 responses counteracted by PPARγ agonists. Theranostics. 2021;11:4531–4548. doi: 10.7150/thno.54803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Du H, Jia A, Gu M, Li S, Zhang P, Bai J. Thyrotoxicosis and osteomalacia: from symptom to pathogenesis. Am J Transl Res. 2021;13:803–811. [PMC free article] [PubMed] [Google Scholar]
  • 135.Cao T, Tang M, Jiang P, Zhang B, Wu X, Chen Q, Zeng C, Li N, Zhang S, Cai H. A potential mechanism underlying the therapeutic effects of progesterone and allopregnanolone on ketamine-induced cognitive deficits. Front Pharmacol. 2021;12:612083. doi: 10.3389/fphar.2021.612083. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES