Current treatments for advanced melanoma and introduction of a promising novel gene therapy for melanoma (Review)

  • Authors:
    • Jae‑Rim Heo
    • Nam‑Hyung Kim
    • Jaejin Cho
    • Kyung‑Chul Choi
  • View Affiliations

  • Published online on: August 24, 2016     https://doi.org/10.3892/or.2016.5032
  • Pages: 1779-1786
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Metastatic melanoma is a fatal form of skin cancer that has a tendency to proliferate more rapidly than any other solid tumor. Since 2010, treatment options for metastatic melanoma have been developed including chemotherapies, checkpoint inhibition immunotherapies, e.g., anti‑cytotoxic T‑lymphocyte antigen‑4 (CTLA‑4) and anti‑programmed death‑1 (PD‑1), and molecular-targeted therapies, e.g., BRAF and MEK inhibitors. These treatments have shown not only high response rates yet also side‑effects and limitations. Notwithstanding its limitations, stem cell therapy has emerged as a new auspicious therapy for various tumor types. Since stem cells possess the ability to serve as a novel vehicle for delivering therapeutic or suicide genes to primary or metastatic cancer sites, these cells can function as part of gene‑directed enzyme prodrug therapy (GDEPT). This review focuses on introducing engineered neural stem cells (NSCs), which have tumor‑tropic behavior that allows NSCs to selectively approach primary and invasive tumor foci, as a potential gene therapy for melanoma. Therapy using engineered NSCs with cytotoxic agents resulted in markedly reduced tumor volumes and significantly prolonged survival rates in preclinical models of various tumor types. This review elucidates current treatment options for metastatic melanoma and introduces a promising NSC therapy.

1. Introduction

Melanoma is the most dangerous form of skin cancer. These invasive growths develop when unrepaired DNA damage causes mutations in skin cells resulting in prompt proliferation and the formation of malignant tumors. If melanoma is detected early, it can be easily cured with appropriate treatment such as surgical removal. However, metastatic melanoma often proves fatal and certain patients possess high-risk features for developing metastases. Melanoma can metastasize almost anywhere, from nearby tissues to distant major organs. The most typical metastatic sites are the lymph nodes, lungs, liver, brain and bones. Many academic reports have been published since 2010 on treatments for metastatic melanoma, from chemotherapies to molecular-targeted therapies.

Research concerning the application of stem cell-based therapies for cancer has recently emerged due to their potential function as a drug delivery vehicle for therapeutic genes directly to tumor sites. Stem cells, such as mesenchymal stem cells (MSCs) and neural stem cells (NSCs), are attractive delivery systems, as they are able to target tumor sites specifically due to the secretion of chemoattractant factors from tumors. Their ability to migrate and aggregate around the tumor at a high concentration gives them the potential as a vector of enzyme/prodrug gene in gene-directed enzyme prodrug therapy (GDEPT) of human cancers (1). This review discusses the current treatment options for metastatic melanoma patients and elucidates the possibility of applying NSC therapy for melanoma and justifying it with prior research on other stem cell-based therapies for melanoma.

2. Current treatments for advanced melanoma

Surgery

Identifying melanoma in its early stages is extremely important since patients with early stage melanoma can be surgically healed with relatively limited associated morbidity (2). Increasing patient survival can be accomplished by accompanying effective palliative management of local disease with removal of systemic, especially solitary lung, melanoma metastases (3). Of 144 patients who underwent surgical resection of non-regional melanoma metastases, 20% had a 5-year survival rate (4), and in a phase II trial conducted by the Southwest Oncology Group the overall 3- and 4-year survival rates of stage IV melanoma patients were 36 and 31%, respectively (5). Surgical tumor removal can prevent metastasis; however, surgical removal cannot be applied on microscopic metastases. Therefore, it must be used with other therapies such as surgical resection concomitant with systemic targeted therapies.

Radiation therapy

A total of 1–6% of patients with melanoma undergo radiation therapy in the USA. In particular, radiation therapy is used in patients with brain metastases as adjunct palliative therapy. Radiation therapy, in contrast to surgical management, has the benefit of potentially inducing an abscopal effect in which both the treated tumor and the non-irradiated site respond to the therapy (2). This abscopal effect is believed to be generated through immune system mediation, as radiation therapy can induce cross-priming in which released tumor antigens are expressed in MHC class I molecules by dendritic cells. Activated CD8+ T cells can then migrate to far-off tumors and promote lysis (6).

Chemotherapy

Dacarbazine (DTIC) is well known as a primary chemotherapeutic treatment for metastatic melanoma. It is the first and only alkylating agent approved by the FDA with intravenous administration every 3–4 weeks at a dose of 800–1,000 mg/m2 (7). DTIC functions by adding an alkyl group to the bases in DNA, which then prevents cells from replicating. As the sole agent of treatment, DTIC creates a partial response in up to 25% of melanomas and a complete response in ~5% (8,9). Oral delivery of the DTIC derivative, temozolamide, which showed a similar response rate in metastatic melanoma, was developed more recently. Temozolamide has the added ability to cross the blood-brain barrier, which led to it becoming a first-line therapy for brain metastases (10). Analysis of combinations of chemotherapies such as cisplatin, vinblastine, and DTIC has shown encouraging response rates, but they have failed to prolong overall survival (OS) when compared with the single agent DTIC (11,12).

Immunotherapy
Cytotoxic T-lymphocyte antigen-4 (CTLA-4)

Activated T cells express CTLA-4. This acts as a negative regulator of T cells and helps preserve immunologic homeostasis. Ipilimumab is an antibody that blocks CTLA-4 from mediating T-cell downregulation and reinforces the antitumor effects of T cells (13). Response rates for ipilimumab alone range from 5 to 15% due to changes in the dosage and patient selection in clinical trials (1417). A total of 1,861 patients were analyzed in 12 separate studies, and the median OS was 11.4 months (95% CI, 10.7–12.1 months) with a plateau at 22% in the survival curve around year 3 (18). Unfortunately, diarrhea, dermatitis, hepatitis, endocrinopathies, and immune-related adverse events accompanied the treatment (19). Clinical trials which focused on a combination of immunotherapy and chemotherapy such as ipilimumab and DTIC exhibited greater efficacy than monotherapy. Patients treated with both ipilimumab and DTIC showed higher OS than those who were treated with only DTIC. The estimated survival rates for patients treated with a combination therapy of ipilimumab and DTIC were 47.3% for 1 year, 28.5% for 2 years, and 20.8% for 3 years compared to survival rates of 36.3, 17.9 and 12.2% for patients treated with DTIC monotherapy (14). Adverse effects of ipilimumab such as gastrointestinal perforations, diarrhea and colitis were less common in groups treated with a combination of ipilimumab and DTIC rather than ipilimumab alone at the same dose, but there were reports of elevated liver function values (14).

Programmed death-1 (PD-1)

T cells upregulate a surface receptor called PD-1 at later stages of T-cell activation in contrast to CTLA-4, which is upregulated in the early stages of T-cell activation. PD-1 regulates the immune system by binding to T cells and attenuating their activity. Tumors are thought to avoid an immune response by upregulating PD-LI, a ligand of PD-1 (20,21). Therefore, preventing the PD-1 ligand from binding to the PD-1 receptor on tumor cells can recover the tumor-fighting function of immune cells. Nivolumab and pembrolizumab are antagonists of the PD-1 receptor that can disrupt the interaction of PD-1 and PD-L1. This disruption can allow T cells to proliferate, infiltrate the tumor and increase effector function (22). Pembrolizumab showed a 38% response rate with median survival of >7 months in initial clinical trials. In comparison with other melanoma treatments, the side-effects were significantly diminished (14,23). Nivolumab had parallel results for the treatment of ipilumumab-resistant or BRAF inhibitor and ipilimumab-resistant advanced melanoma. With nivolumab, 31.7% of patients had an objective response compared to 10.6% of patients who were treated with the investigator's choice of chemotherapy (ICC) (24). Adopting a combination therapy of nivolumab and ipilimumab has shown the highest response rates. Using these therapies simultaneously in phase I and II trials demonstrated a 53–61% response rate with >80% tumor reduction in all responding patients (25,26). This result shows a synergistic effect between CTLA-4 and PD-1 inhibition and a recent report [Larkin et al (2015)] showed that the median progression-free survival (PFS) of the combination therapy was 11.5 months (95% CI, 8.9–16.7) compared with 2.9 months (95% CI, 2.8–3.4) with ipilimumab alone and 6.9 months (95% CI, 4.3–9.5) with nivolumab alone (27). Vitiligo, colitis, hepatitis, hypophysitis, and thyroiditis are adverse events of nivolumab, but they appear less often compared to the treatment with a CTLA-4 antagonist. In addition, inflammatory pneumonitis along with a dry cough, dyspnea, and ground opacities are unique to PD-1 blockade and are potentially lethal (28).

Molecular-targeted therapy
BRAF inhibitor

The BRAF gene encodes a serine/threonine kinase that is engaged in the mitogen-activated protein kinase (MAPK)/ERK signaling pathway (29). The MAPK/ERK signaling pathway is associated in transferring signals for cellular proliferation and survival from the cell surface to the nucleus, and ~50% of cutaneous melanomas are caused by a mutation in the BRAF oncogene, which leads to fundamental activation of the MAPK signaling pathway and uncontrolled cellular proliferation (30,31). Vemurafenib and dabrafenib are potent BRAF inhibitors with distinct antitumor effects specific to melanoma cell lines with the BRAF V600E and V600E/K mutations (3235). In its initials trials, treatment with vemurafenib induced complete or partial tumor regression in 81% of patients with melanoma containing the V600E BRAF mutation (32). Vemurafenib received approval for BRAF inhibitor monotherapy in 2011. Dabrafenib is another BRAF-targeted therapy for melanoma which functions as a reversible ATP-competitive inhibitor for BRAF and was approved in March 2013 (31). Median PFS for vemurafenib is 6.8 months compared to 5.1 months for dabrafenib, which signifies that dabrafenib is not more effective than vemurfaneib monotherapy (34,36). However, a study showed that dabrafenib demonstrated efficacy for patients with brain metastases and remains an effective therapeutic option for this particular population (37). Vemurafenib showed favorable in vitro and in vivo results and a 69% objective response rate in phase I clinical trials (38,39). As clinical trials proceeded to later phases, however, 90% of patients gained resistance and showed disease progression within 9 months.

Arthralgia, fatigue, aminotransferase elevation, nausea, vomiting and decreased kidney function were reported as general side-effects of vemurafenib, and ~11% of patients administered dabrafenib reported pyrexia as a side-effect (13,34).

BRAF resistance

Repeated exposure to mutant BRAF inhibitors can alter not only the RAS-RAF-MEK-ERK signaling pathway but also several other kinase pathways (36). As a result, expression levels of RAS, CRAF and MEK were increased due to ERK pathway reactivation (40,41). ERK signaling reactivation is driven by the amplification or alternative splicing of BRAF causing BRAF dimerization that prevents inhibitors from binding to BRAF V600E monomers (42,43). For example, activation of the PI3K/AKT signaling pathway promotes BRAF inhibitor resistance in melanoma and is therefore a form of adaptive resistance (44). Changes in the tumor microenvironment caused by increased levels of growth factors such as hepatocyte growth factor (HGF) can be another mechanism for BRAF inhibitor resistance and were found to be linked to poor clinical outcomes (45,46). However, changes in tumor microenvironment are not hypothesized to be the primary cause of drug resistance, but they are considered to be a secondary contributor which could be a targetable option for preventing adaptive resistance in melanoma tumors (47).

MEK inhibitor

While direct targeting of mutated oncogenic BRAF has been successful for those with mutated BRAF metastatic melanoma, blocking MEK, a protein located downstream of BRAF in the MAPK signaling pathway, showed remarkable success as well. Compared to oncogenic BRAF mutations, oncogenic MEK mutations are less common in melanoma. However, because of BRAF inhibitor resistance, targeting downstream of BRAF for therapeutic efficacy has become a research topic of interest (13,31). The common MEK mutation C121S accelerates melanoma growth and confers resistance to BRAF V600E mutant melanoma cells to vemurafenib. C121S creates an active kinase that allows for activation of downstream ERK without upstream activation by BRAF (48). A MEK inhibitor called trametinib has been FDA approved as a single agent for melanoma patients with BRAF V600E or V600K mutations as of June 2013 (13). Trametinib impedes the progression of advanced melanoma, especially in BRAF-mutant patients (17,49). Trametinib showed a 33% response rate for BRAF mutants with 5.6 months of median PFS in recent clinical trials compared to a 10% response rate for BRAF wild-type tumors (50,51). Although trametinib showed more improvement in PFS and OS compared with chemotherapy, the objective response rate was still lower than that of BRAF inhibitors (13). Furthermore, trametinib produced side-effects including diarrhea, peripheral edema, hypertension and fatigue, which are typical of other MEK inhibitors as well (52). Many resistance pathways found in other treatments, especially BRAF inhibitors, depend upon MEK signaling. Thus, MEK inhibition by trametinib in combination with other treatments was able to increase their potential as therapeutic agents and attenuate resistance in clinical trials (50,51).

Combination-targeted therapy

BRAF resistance from BRAF kinase inhibitors is generated by reactivation of the MAPK pathway. In order to solve this problem, Flaherty et al performed a combined treatment with a selective BRAF inhibitor, dabrafenib, and a selective MEK inhibitor, trametinib, in phase I and II trials (53). Vemurafenib was found to inhibit MAPK signaling in melanoma patients with the BRAF V600E mutation and produce prolonged survival and PFS in randomized phase III trials in patients who had not previously received melanoma treatments. Trametinib restricts MEK, a protein downstream of BRAF in the MAPK pathway, and it showed an improvement in progression-free survival and OS in BRAF V600E and V600K mutant melanomas. Rapid reactivation of the MAPK signaling pathway has been related to BRAF inhibitor resistance in preclinical models, but stimulation of cell death in BRAF V600 mutant melanoma requires complete inhibition of the MAPK pathway. This can be attained by combining a BRAF inhibitor with an MEK inhibitor (53). The median OS for combined treatment with trametinib and dabrafenib in a multicenter, double-blind, phase III randomized controlled trial on BRAF-mutant melanoma patients was 25.1 months (95% CI, 19.2-not reached) and 18.7 months (15.2–23.7) for the dabrafenib only-treated BRAF-mutant melanoma patients [hazard ratio (HR), 0.71; 95% CI, 0.55–0.92; p=0.0107]. Median PFS for the dabrafenib and trametinib-combined therapy was 11.0 months (95% CI, 8.0–13.9) and for the dabrafenib only-treated group this value was 8.8 months (5.5–9.3) (HR, 0.67; 95% CI, 0.53–0.84; p=0.0004) (54). Flaherty et al examined the adverse side-effects of combination therapy with dabrafenib and trametinib. Patients who received both dabrafenib and trametinib treatment had more constant and severe pyrexia and chills compared to those who had only dabrafenib treatment. They also had more persistent gastrointestinal toxic effects, such as nausea and vomiting, but the majority were grade 1 or 2 events (53).

3. A promising novel therapy for cancer - neural stem cell therapy

Neural stem cells

NSCs are self-renewing created by the differentiation of embryonic tissue and generate the neurons and glia of the developing brain. NSCs can be isolated, genetically engineered and differentiated in vitro and reinstated into the central nervous system (CNS). NSCs have potential for use in cell replacement therapies in various neurologic disorders as has been shown in several academic reports (55,56). NSCs can be defined as cells that self-renew constantly and have the potential to form intermediate and mature cells of neuronal and glial lineages (57). From the year 2000 onward, there have been many reports concerning the adoption of NSCs as a drug delivery vehicle specific to brain sites instead of solely cell replacement. NSCs were found to appear near metastatic tumor cells far from where they were transplanted into animal models of brain neoplasia in these reports. This opens the possibility to track down and destroy malignant cells by manufacturing NSCs with chemotherapeutic qualities (5860). NSCs have the unique ability to integrate into the host's brain without interfering with normal functions and can proliferate for long periods (61). This uniqueness could allow NSCs to be suitable as therapeutic delivery vehicles for CNS disorders. In addition, their tropic migration towards neoplasms is another favorable characteristic for their use as vehicles for targeted delivery (55). Benedetti et al and Aboody et al demonstrated that the progression of cancer xenografts was suppressed by the cytotoxic effects of NSCs that were manufactured with antitumor gene products (58,60). These studies opened the doors to the potential of drug-equipped NSCs as a tumor-homing therapy. NSCs migrate not only to injured areas but also towards tumor foci. The tumor-tropic homing of NSCs is directed by chemoattractants produced by cells in the normal brain wounded by tumor growth or directly released from glioblastoma multiforme (GBM) cells (62,63). In hypoxic conditions, GBM cells upregulate the expression of numerous pro-angiogenic factors and chemoattractants. The relevance of hypoxia in the tumor-tropic migration of NSCs towards GBM was demonstrated through several siRNA-mediated knockdowns. The expression of the chemoattractant factor stromal cell-derived factor-1 (SDF-1), uPA and vascular endothelial growth factor (VEGF) was reduced with knockdown of HIG-α in GBM cells, which led to no tumor-tropic migration of NSCs (62). More cytokines, growth factors, and receptors have been addressed such as (SCF)/c-Kit (64), monocyte chemoattractant protein-1 (MCP-1)/CCL2 (65), Annexin A2 (66), HGF/c-Met (67) and HMGB1/RAGE (68) for arbitrating the tumor-tropic migration of NSCs. Engineered NSCs could be designed to express a plurality of receptors, so they can be deployed wherever chemotactic signals are emitted from brain pathologies. Various groups have revealed the potential of migrating towards not only tumors of glial origin but also metastatic breast cancer and melanoma foci in the brain (6971). Due to their intrinsic migratory and tumor-tropic properties, NSCs epitomize a novel and potentially efficacious approach for the treatment of invasive tumors.

Gene-directed enzyme prodrug therapy of human cancer

Conventional treatments of cancer are impeded by their inadequacy in being selective and specific to de novo tumors. They harm normal and healthy tissues by their toxicity. HB1.F3 cells, a parental cell line of the HB1.F3.CD/CE cell line, show migration to subcutaneous xenografts of various solid tumors such as prostate cancer, breast cancer, melanoma, glioma and neuroblastoma. This suggests that these cell lines do not show tissue-specific characteristics for therapeutic use (70).

GDEPT is a promising approach for advancing the selectivity of conventional chemotherapeutics. GDEPT improves selectivity by delivering 'suicide' genes such as cytosine deaminase (CD), carboxylesterase (CE), and herpes simplex virus type 1 thymidine kinase (HSV1-tk), to cancer cells, which lets them convert non- or low-cytotoxic prodrugs to cytotoxic drugs (58,72,73). Using GDEPT allows human tumors to be selectively targeted and specifically treated to increase efficacy and diminish the side-effects of biological drugs (74). For example, CD converts 5-fluorocytosine (5-FC), a non-toxic drug, to 5-fluorouracil (5-FU), a toxic agent, CE converts CPT-11 to SN-38, and HSV1-tk converts GCV to an active metabolite. An essential aspect of GDEPT is a foreign enzyme expressed only at the tumor site where it is able to shift a prodrug into its cytotoxic metabolite in vivo (58). The therapeutic efficacy of a polymerase chain reaction (PCR) vector which conveyed a suicide gene, yeast CD, that converts the prodrug 5-FC to the cytotoxic 5-FU was exhibited after delivery by infusion into the regional circulation in a multifocal hepatic metastasis model of colon cancer (75). A noticeable boost in apoptotic cells and a decrease in proliferated cells in human breast cancer cell lines was detected when combined treatment was used with the CD/5-FC suicide system and hTNFα expression (76).

Tropism of neural stem/progenitor cells to human cancers

Selective penetration to tumor sites is the primary handicap that current gene therapy strategies are confronting, but this can be overcome by using NSCs. NSCs are able to serve as a delivery vehicle to target and propagate therapeutic gene products over tumor sites. The human NSC line HB1.F3.CD was implanted intracranially at distant sites from the tumor, and the NSCs selectively migrated to the GBM tumor mass while bypassing normal tissue which resulted in 80–85% reduction in tumor volume after injection of the prodrug 5-FC (69,77,78). NSCs are assumed to have a bystander effect through their selectively eliminating behavior against dividing tumor cells wherein toxic prodrugs and their metabolites circulate across gap junctions and interstitial space to surrounding cells (74). Although the selective migration towards tumor sites of HB1.F3 parental cells, the HB1.F3.CD/CE cell line and other stem/progenitor cells has not been fully explained, biological factors such as SDF-1, scatter factor (SCF; HGF), VEGF and MCP-1 expressed in tumor cells seem to participate in chemotaxis to human tumors (55,64,68,7983). Adopting the tumor-tropic behavior of NSCs could lead to significant utility for the treatment of a variety of metastatic tumors.

Alternative stem cell-based therapies for melanoma treatment

Among many types of stem cells, MSCs have emerged as a potential transporter for not only regenerative medicine but also cancer therapy. There have been several studies suggesting that MSCs are able to migrate to both primary and metastatic tumor sites through associations with various chemokines and cytokines (1). Similar to NSCs, MSCs can track specifically to tumor sites via chemokines and cytokines emitted from tumors (8487). There is a large body of research concerning the application of MSCs as carriers of anticancer agents for melanoma treatments. For example, bone marrow-derived MSCs engineered to carry the P450 gene showed the ability to inhibit the growth of malignant melanoma in vitro and in vivo by reinforcing the expression of CYP2E1 (88). A study by Jing et al used adipose tissue-derived mesenchymal stromal cells (AT-MSCs) as a carrier to deliver enhanced expression of TRAIL protein for impeding melanoma growth. TRAIL protein induced apoptosis by readjusting the expression of members of the PI3K-AKT signaling pathway (89). Seo et al demonstrated the antitumor effect of engineered canine AT-MSC (cAT-MSC)-producing interferon-β with cisplatin in mouse melanoma models. The combination of cAT-MSC-IFN-β and cisplatin had more compelling results than the cisplatin-alone group in inhibiting the growth of melanoma and increasing the survival rate (90). Tyciakova et al used engineered AT-MSC-secreting TNFα protein to assess its therapeutic effect on melanoma. AT-MSC-TNFα restrained melanoma cells from growth in vitro by inducing apoptosis via activating caspase-3/7 and inhibited the tumor mass up to 97.5% (91). All these studies suggest that stem cells are satisfactory as a carrier of both anticancer drugs and genes for targeting cancers. Overall, the data obtained from alternative stem cell-based therapies on melanoma propose the feasibility of NSCs as a delivery system for targeted agents in the treatment of melanoma.

4. Conclusions

As we learn more about the mechanisms of melanoma, treatment has been revolutionized. The advancement of immunotherapies and targeted therapies has significantly improved clinical results. However, although there are currently more wide-ranging treatment options than in the past, it has become apparent that monotherapy will likely be unsuccessful due to the aggressiveness and hypermutable nature of melanoma tumors. Thus far, combination therapy has produced the most convincing clinical results. Although both immunotherapy and targeted therapies have conspicuous advantages and disadvantages, preclinical results show that the combination of these treatments could enhance patient outcomes. However, related data are inadequate to make a concrete determination, as the data of patients treated with combination therapy are limited. The toxicity and resistance issues plaguing many existing treatments must also be carefully considered with combination therapy. Therefore, the ultimate efficacy of combination therapies remains unclear until further data are gathered.

The common obstacle that current melanoma treatment options confront is damage to other tissues. This issue has placed patients in situations where whether to continue their treatment or not has been a serious consideration for maximizing their chances of survival. However, a parental cell line of HB1.F3.CD/CE has been demonstrated to exhibit migratory behavior to subcutaneous xenografts of various solid tumors in the prostate and breast as well as melanoma, glioma, and neuroblastoma (Fig. 1). We can interpret that engineered NSCs with suicide genes can be used to selectively target not only melanoma but also tumors that have already metastasized to other sites without damaging normal tissues for therapeutic use, as demonstrated in Table I. Although, to date, research is lacking regarding the use of engineered NSCs for melanoma, data from other stem cell-based therapies on melanoma and the features of NSCs indicate that NSC therapy could be the next paradigm in gene therapy for melanoma and other cancers in preclinical and clinical cases. Thus, their potential as a specialized delivery vehicle should be explored in future studies.

Table I

Engineered stem cells for therapeutic efficacy in preclinical models of different tumor types.

Table I

Engineered stem cells for therapeutic efficacy in preclinical models of different tumor types.

Tumor type (cell line)NSC type (prodrug)Tumor volumea (%)Survival rateRef.
Breast cancer (MDA-MB-231/luc)HB1.F3.CD (5-FC)5030% (13 weeks)(92)
HB1.F3.CD.IFN-β (5-FC)5080% (14 weeks)
Endometrial cancer (Ishikawa)HB1.F3.CD (5-FC)50NDb(93)
HB1.F3.CD.IFN-β (5-FC)60NDb
Human colorectal cancer (HT-29)HB1.F3.CD (5-FC)5650% (11 weeks)(94)
HB1.F3.CD.IFN-β (5-FC)7670% (11 weeks)
Pancreatic cancer (PANC-1)HB1.F3.CD (5-FC)50NDb(95)
HB1.F3.CD.IFN-β (5-FC)50NDb
Lung cancer (A549)HB1.F3.CE (CPT-11)80NDb(96)

a Tumor volume is shown in a ratio of negative control to treatment group.

b ND, no data are shown. NSC, neural stem cell; 5-FC, 5-fluorocytosine.

Acknowledgments

This study was supported by the Basic Science Research Program through the National Research Foundation (NRF) of Korea funded by the Ministry of Education, Science and Technology (MEST) (2013R1A1A2059092). In addition, this study was supported by a grant from the Next-Generation BioGreen 21 Program (no. PJ011355), Rural Development Administration, Republic of Korea.

References

1 

Mirzaei H, Sahebkar A, Avan A, Jaafari MR, Salehi R, Salehi H, Baharvand H, Rezaei A, Hadjati J, Pawelek JM, et al: Application of mesenchymal stem cells in melanoma: A potential therapeutic strategy for delivery of targeted agents. Curr Med Chem. 23:455–463. 2016. View Article : Google Scholar

2 

Maverakis E, Cornelius LA, Bowen GM, Phan T, Patel FB, Fitzmaurice S, He Y, Burrall B, Duong C, Kloxin AM, et al: Metastatic melanoma - a review of current and future treatment options. Acta Derm Venereol. 95:516–524. 2015. View Article : Google Scholar

3 

Younes R, Abrao FC and Gross J: Pulmonary metastasectomy for malignant melanoma: Prognostic factors for long-term survival. Melanoma Res. 23:307–311. 2013.PubMed/NCBI

4 

Wong JH, Skinner KA, Kim KA, Foshag LJ and Morton DL: The role of surgery in the treatment of nonregionally recurrent melanoma. Surgery. 113:389–394. 1993.PubMed/NCBI

5 

Sosman JA, Moon J, Tuthill RJ, Warneke JA, Vetto JT, Redman BG, Liu PY, Unger JM, Flaherty LE and Sondak VK: A phase 2 trial of complete resection for stage IV melanoma: Results of Southwest Oncology Group Clinical Trial S9430. Cancer. 117:4740–4706. 2011. View Article : Google Scholar : PubMed/NCBI

6 

National Cancer Institute of Canada Melanoma Group: Vinblastine, bleomycin, and cis-platinum for the treatment of metastatic malignant melanoma. J Clin Oncol. 2:131–134. 1984.

7 

Kim T, Amaria RN, Spencer C, Reuben A, Cooper ZA and Wargo JA: Combining targeted therapy and immune checkpoint inhibitors in the treatment of metastatic melanoma. Cancer Biol Med. 11:237–246. 2014.

8 

Eggermont AM and Kirkwood JM: Re-evaluating the role of dacarbazine in metastatic melanoma: What have we learned in 30 years? Eur J Cancer. 40:1825–1836. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Huncharek M, Caubet JF and McGarry R: Single-agent DTIC versus combination chemotherapy with or without immunotherapy in metastatic melanoma: A meta-analysis of 3273 patients from 20 randomized trials. Melanoma Res. 11:75–81. 2001. View Article : Google Scholar : PubMed/NCBI

10 

Middleton MR, Grob JJ, Aaronson N, Fierlbeck G, Tilgen W, Seiter S, Gore M, Aamdal S, Cebon J, Coates A, et al: Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma. J Clin Oncol. 18:158–166. 2000.PubMed/NCBI

11 

Chapman PB, Einhorn LH, Meyers ML, Saxman S, Destro AN, Panageas KS, Begg CB, Agarwala SS, Schuchter LM, Ernstoff MS, et al: Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J Clin Oncol. 17:2745–2751. 1999.PubMed/NCBI

12 

Legha SS, Ring S, Papadopoulos N, Plager C, Chawla S and Benjamin R: A prospective evaluation of a triple-drug regimen containing cisplatin, vinblastine, and dacarbazine (CVD) for metastatic melanoma. Cancer. 64:2024–2029. 1989. View Article : Google Scholar : PubMed/NCBI

13 

A Schindler K and Postow MA: Current options and future directions in the systemic treatment of metastatic melanoma. J Community Support Oncol. 12:20–26. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Robert C, Thomas L, Bondarenko I, O'Day S, Weber J, Garbe C, Lebbe C, Baurain JF, Testori A, Grob JJ, et al: Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 364:2517–2526. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Hersh EM, O'Day SJ, Powderly J, Khan KD, Pavlick AC, Cranmer LD, Samlowski WE, Nichol GM, Yellin MJ and Weber JS: A phase II multicenter study of ipilimumab with or without dacarbazine in chemotherapy-naïve patients with advanced melanoma. Invest New Drugs. 29:489–498. 2011. View Article : Google Scholar

16 

Kaplan MG: Ipilimumab plus dacarbazine in melanoma. N Engl J Med. 365:1256–1257; author reply 1257–1258. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Luke JJ, Callahan MK, Postow MA, Romano E, Ramaiya N, Bluth M, Giobbie-Hurder A, Lawrence DP, Ibrahim N, Ott PA, et al: Clinical activity of ipilimumab for metastatic uveal melanoma: A retrospective review of the Dana-Farber Cancer Institute, Massachusetts General Hospital, Memorial Sloan-Kettering Cancer Center, and University Hospital of Lausanne experience. Cancer. 119:3687–3695. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O, Patt D, Chen TT, Berman DM and Wolchok JD: Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol. 33:1889–1894. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Weber JS, Kähler KC and Hauschild A: Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol. 30:2691–2697. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Zou W and Chen L: Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 8:467–477. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Keir ME, Liang SC, Guleria I, Latchman YE, Qipo A, Albacker LA, Koulmanda M, Freeman GJ, Sayegh MH and Sharpe AH: Tissue expression of PD-L1 mediates peripheral T cell tolerance. J Exp Med. 203:883–895. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, et al: PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 515:568–571. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, et al: Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 369:134–144. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Weber JS, D'Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, Hoeller C, Khushalani NI, Miller WH Jr, Lao CD, et al: Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): A randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 16:375–384. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K, et al: Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 369:122–133. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, Linette GP, Meyer N, Giguere JK, Agarwala SS, et al: Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 372:2006–2017. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Larkin J, Hodi FS and Wolchok JD: Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 373:1270–1271. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al: Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 366:2443–2454. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, et al: Mutations of the BRAF gene in human cancer. Nature. 417:949–954. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Davies MA, Stemke-Hale K, Lin E, Tellez C, Deng W, Gopal YN, Woodman SE, Calderone TC, Ju Z, Lazar AJ, et al: Integrated molecular and clinical analysis of AKT activation in metastatic melanoma. Clin Cancer Res. 15:7538–7546. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Davey RJ, van der Westhuizen A and Bowden NA: Metastatic melanoma treatment: Combining old and new therapies. Crit Rev Oncol Hematol. 98:242–253. 2016. View Article : Google Scholar

32 

Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, O'Dwyer PJ, Lee RJ, Grippo JF, Nolop K, et al: Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med. 363:809–819. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Ribas A and Flaherty KT: BRAF targeted therapy changes the treatment paradigm in melanoma. Nat Rev Clin Oncol. 8:426–433. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Hauschild A, Grob JJ, Demidov LV, Jouary T, Gutzmer R, Millward M, Rutkowski P, Blank CU, Miller WH Jr, Kaempgen E, et al: Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet. 380:358–365. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Bollag G, Hirth P, Tsai J, Zhang J, Ibrahim PN, Cho H, Spevak W, Zhang C, Zhang Y, Habets G, et al: Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature. 467:596–599. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Sosman JA, Kim KB, Schuchter L, Gonzalez R, Pavlick AC, Weber JS, McArthur GA, Hutson TE, Moschos SJ, Flaherty KT, et al: Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N Engl J Med. 366:707–714. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Long GV, Trefzer U, Davies MA, Kefford RF, Ascierto PA, Chapman PB, Puzanov I, Hauschild A, Robert C, Algazi A, et al: Dabrafenib in patients with Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain (BREAK-MB): A multicentre, open-label, phase 2 trial. Lancet Oncol. 13:1087–1095. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Heakal Y, Kester M and Savage S: Vemurafenib (PLX4032): An orally available inhibitor of mutated BRAF for the treatment of metastatic melanoma. Ann Pharmacother. 45:1399–1405. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Luke JJ and Hodi FS: Vemurafenib and BRAF inhibition: A new class of treatment for metastatic melanoma. Clin Cancer Res. 18:9–14. 2012. View Article : Google Scholar

40 

Lo RS and Shi H: Detecting mechanisms of acquired BRAF inhibitor resistance in melanoma. Methods Mol Biol. 1102:163–174. 2014. View Article : Google Scholar

41 

Sullivan RJ and Flaherty KT: Resistance to BRAF-targeted therapy in melanoma. Eur J Cancer. 49:1297–1304. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Poulikakos PI, Persaud Y, Janakiraman M, Kong X, Ng C, Moriceau G, Shi H, Atefi M, Titz B, Gabay MT, et al: RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E). Nature. 480:387–390. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Shi H, Hugo W, Kong X, Hong A, Koya RC, Moriceau G, Chodon T, Guo R, Johnson DB, Dahlman KB, et al: Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy. Cancer Discov. 4:80–93. 2014. View Article : Google Scholar :

44 

Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga-Kalabis M, Cipolla AK, Wubbenhorst B, Xu X, Gimotty PA, Kee D, et al: Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell. 18:683–695. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Koefinger P, Wels C, Joshi S, Damm S, Steinbauer E, Beham-Schmid C, Frank S, Bergler H and Schaider H: The cadherin switch in melanoma instigated by HGF is mediated through epithelial-mesenchymal transition regulators. Pigment Cell Melanoma Res. 24:382–385. 2011. View Article : Google Scholar :

46 

Topcu-Yilmaz P, Kiratli H, Saglam A, Söylemezoglu F and Hascelik G: Correlation of clinicopathological parameters with HGF, c-Met, EGFR, and IGF-1R expression in uveal melanoma. Melanoma Res. 20:126–132. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Gajewski TF: Identifying and overcoming immune resistance mechanisms in the melanoma tumor microenvironment. Clin Cancer Res. 12:2326s–2330s. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Wagle N, Emery C, Berger MF, Davis MJ, Sawyer A, Pochanard P, Kehoe SM, Johannessen CM, Macconaill LE, Hahn WC, et al: Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling. J Clin Oncol. 29:3085–3096. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Kim KB, Kefford R, Pavlick AC, Infante JR, Ribas A, Sosman JA, Fecher LA, Millward M, McArthur GA, Hwu P, et al: Phase II study of the MEK1/MEK2 inhibitor Trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitor. J Clin Oncol. 31:482–489. 2013. View Article : Google Scholar

50 

Menzies AM and Long GV: Dabrafenib and trametinib, alone and in combination for BRAF-mutant metastatic melanoma. Clin Cancer Res. 20:2035–2043. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Kim MO, Kim SH, Oi N, Lee MH, Yu DH, Kim DJ, Cho EJ, Bode AM, Cho YY, Bowden TG, et al: Embryonic stem-cell-preconditioned microenvironment induces loss of cancer cell properties in human melanoma cells. Pigment Cell Melanoma Res. 24:922–931. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Kirkwood JM, Bastholt L, Robert C, Sosman J, Larkin J, Hersey P, Middleton M, Cantarini M, Zazulina V, Kemsley K, et al: Phase II, open-label, randomized trial of the MEK1/2 inhibitor selumetinib as monotherapy versus temozolomide in patients with advanced melanoma. Clin Cancer Res. 18:555–567. 2012. View Article : Google Scholar

53 

Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, Hamid O, Schuchter L, Cebon J, Ibrahim N, et al: Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med. 367:1694–1703. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, Garbe C, Jouary T, Hauschild A, Grob JJ, et al: Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: A multicentre, double-blind, phase 3 randomised controlled trial. Lancet. 386:444–451. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Müller FJ, Snyder EY and Loring JF: Gene therapy: Can neural stem cells deliver? Nat Rev Neurosci. 7:75–84. 2006. View Article : Google Scholar

56 

Bago JR, Sheets KT and Hingtgen SD: Neural stem cell therapy for cancer. Methods. 99:37–43. 2016. View Article : Google Scholar

57 

Gage FH: Mammalian neural stem cells. Science. 287:1433–1438. 2000. View Article : Google Scholar : PubMed/NCBI

58 

Benedetti S, Pirola B, Pollo B, Magrassi L, Bruzzone MG, Rigamonti D, Galli R, Selleri S, Di Meco F, De Fraja C, et al: Gene therapy of experimental brain tumors using neural progenitor cells. Nat Med. 6:447–450. 2000. View Article : Google Scholar : PubMed/NCBI

59 

Herrlinger U, Woiciechowski C, Sena-Esteves M, Aboody KS, Jacobs AH, Rainov NG, Snyder EY and Breakefield XO: Neural precursor cells for delivery of replication-conditional HSV-1 vectors to intracerebral gliomas. Mol Ther. 1:347–357. 2000. View Article : Google Scholar : PubMed/NCBI

60 

Aboody KS, Brown A, Rainov NG, Bower KA, Liu S, Yang W, Small JE, Herrlinger U, Ourednik V, Black PM, et al: Neural stem cells display extensive tropism for pathology in adult brain: Evidence from intracranial gliomas. Proc Natl Acad Sci USA. 97:12846–12851. 2000. View Article : Google Scholar : PubMed/NCBI

61 

Consiglio A, Gritti A, Dolcetta D, Follenzi A, Bordignon C, Gage FH, Vescovi AL and Naldini L: Robust in vivo gene transfer into adult mammalian neural stem cells by lentiviral vectors. Proc Natl Acad Sci USA. 101:14835–14840. 2004. View Article : Google Scholar : PubMed/NCBI

62 

Zhao D, Najbauer J, Garcia E, Metz MZ, Gutova M, Glackin CA, Kim SU and Aboody KS: Neural stem cell tropism to glioma: Critical role of tumor hypoxia. Mol Cancer Res. 6:1819–1829. 2008. View Article : Google Scholar : PubMed/NCBI

63 

Zhang S, Luo X, Wan F and Lei T: The roles of hypoxia-inducible factors in regulating neural stem cells migration to glioma stem cells and determinating their fates. Neurochem Res. 37:2659–2666. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Sun L, Lee J and Fine HA: Neuronally expressed stem cell factor induces neural stem cell migration to areas of brain injury. J Clin Invest. 113:1364–1374. 2004. View Article : Google Scholar : PubMed/NCBI

65 

Magge SN, Malik SZ, Royo NC, Chen HI, Yu L, Snyder EY, O'Rourke DM and Watson DJ: Role of monocyte chemoattractant protein-1 (MCP-1/CCL2) in migration of neural progenitor cells toward glial tumors. J Neurosci Res. 87:1547–1555. 2009. View Article : Google Scholar : PubMed/NCBI

66 

An JH, Lee SY, Jeon JY, Cho KG, Kim SU and Lee MA: Identification of gliotropic factors that induce human stem cell migration to malignant tumor. J Proteome Res. 8:2873–2881. 2009. View Article : Google Scholar : PubMed/NCBI

67 

Heese O, Disko A, Zirkel D, Westphal M and Lamszus K: Neural stem cell migration toward gliomas in vitro. Neuro Oncol. 7:476–484. 2005. View Article : Google Scholar : PubMed/NCBI

68 

Schmidt NO, Przylecki W, Yang W, Ziu M, Teng Y, Kim SU, Black PM, Aboody KS and Carroll RS: Brain tumor tropism of transplanted human neural stem cells is induced by vascular endothelial growth factor. Neoplasia. 7:623–629. 2005. View Article : Google Scholar : PubMed/NCBI

69 

Kim SK, Kim SU, Park IH, Bang JH, Aboody KS, Wang KC, Cho BK, Kim M, Menon LG, Black PM, et al: Human neural stem cells target experimental intracranial medulloblastoma and deliver a therapeutic gene leading to tumor regression. Clin Cancer Res. 12:5550–5556. 2006. View Article : Google Scholar : PubMed/NCBI

70 

Joo KM, Park IH, Shin JY, Jin J, Kang BG, Kim MH, Lee SJ, Jo MY, Kim SU and Nam DH: Human neural stem cells can target and deliver therapeutic genes to breast cancer brain metastases. Mol Ther. 17:570–575. 2009. View Article : Google Scholar : PubMed/NCBI

71 

Aboody KS, Bush RA, Garcia E, Metz MZ, Najbauer J, Justus KA, Phelps DA, Remack JS, Yoon KJ, Gillespie S, et al: Development of a tumor-selective approach to treat metastatic cancer. PLoS One. 1:e232006. View Article : Google Scholar : PubMed/NCBI

72 

Schepelmann S, Ogilvie LM, Hedley D, Friedlos F, Martin J, Scanlon I, Chen P, Marais R and Springer CJ: Suicide gene therapy of human colon carcinoma xenografts using an armed oncolytic adenovirus expressing carboxypeptidase G2. Cancer Res. 67:4949–4955. 2007. View Article : Google Scholar : PubMed/NCBI

73 

Schepelmann S and Springer CJ: Viral vectors for gene-directed enzyme prodrug therapy. Curr Gene Ther. 6:647–670. 2006. View Article : Google Scholar : PubMed/NCBI

74 

Kim SU, Jeung EB, Kim YB, Cho MH and Choi KC: Potential tumor-tropic effect of genetically engineered stem cells expressing suicide enzymes to selectively target invasive cancer in animal models. Anticancer Res. 31:1249–1258. 2011.PubMed/NCBI

75 

Hiraoka K, Kimura T, Logg CR, Tai CK, Haga K, Lawson GW and Kasahara N: Therapeutic efficacy of replication-competent retrovirus vector-mediated suicide gene therapy in a multifocal colorectal cancer metastasis model. Cancer Res. 67:5345–5353. 2007. View Article : Google Scholar : PubMed/NCBI

76 

Pastorakova A, Hlubinova K, Jakubikova J and Altaner C: Combine cancer gene therapy harnessing plasmids expressing human tumor necrosis factor alpha and Herpes simplex thymidine kinase suicide gene. Neoplasma. 53:353–362. 2006.PubMed/NCBI

77 

Brown AB, Yang W, Schmidt NO, Carroll R, Leishear KK, Rainov NG, Black PM, Breakefield XO and Aboody KS: Intravascular delivery of neural stem cell lines to target intracranial and extracranial tumors of neural and non-neural origin. Hum Gene Ther. 14:1777–1785. 2003. View Article : Google Scholar : PubMed/NCBI

78 

Yip S, Aboody KS, Burns M, Imitola J, Boockvar JA, Allport J, Park KI, Teng YD, Lachyankar M, McIntosh T, et al: Neural stem cell biology may be well suited for improving brain tumor therapies. Cancer J. 9:189–204. 2003. View Article : Google Scholar : PubMed/NCBI

79 

Ehtesham M, Yuan X, Kabos P, Chung NH, Liu G, Akasaki Y, Black KL and Yu JS: Glioma tropic neural stem cells consist of astrocytic precursors and their migratory capacity is mediated by CXCR4. Neoplasia. 6:287–293. 2004. View Article : Google Scholar : PubMed/NCBI

80 

Sun L, Hui AM, Su Q, Vortmeyer A, Kotliarov Y, Pastorino S, Passaniti A, Menon J, Walling J, Bailey R, et al: Neuronal and glioma-derived stem cell factor induces angiogenesis within the brain. Cancer Cell. 9:287–300. 2006. View Article : Google Scholar : PubMed/NCBI

81 

Beppu K, Jaboine J, Merchant MS, Mackall CL and Thiele CJ: Effect of imatinib mesylate on neuroblastoma tumorigenesis and vascular endothelial growth factor expression. J Natl Cancer Inst. 96:46–55. 2004. View Article : Google Scholar : PubMed/NCBI

82 

Geminder H, Sagi-Assif O, Goldberg L, Meshel T, Rechavi G, Witz IP and Ben-Baruch A: A possible role for CXCR4 and its ligand, the CXC chemokine stromal cell-derived factor-1, in the development of bone marrow metastases in neuroblastoma. J Immunol. 167:4747–4757. 2001. View Article : Google Scholar : PubMed/NCBI

83 

Kucia M, Reca R, Miekus K, Wanzeck J, Wojakowski W, Janowska-Wieczorek A, Ratajczak J and Ratajczak MZ: Trafficking of normal stem cells and metastasis of cancer stem cells involve similar mechanisms: Pivotal role of the SDF-1-CXCR4 axis. Stem Cells. 23:879–894. 2005. View Article : Google Scholar : PubMed/NCBI

84 

Vicari AP and Caux C: Chemokines in cancer. Cytokine Growth Factor Rev. 13:143–154. 2002. View Article : Google Scholar : PubMed/NCBI

85 

Lazennec G and Richmond A: Chemokines and chemokine receptors: New insights into cancer-related inflammation. Trends Mol Med. 16:133–144. 2010. View Article : Google Scholar : PubMed/NCBI

86 

Loebinger MR and Janes SM: Stem cells as vectors for antitumour therapy. Thorax. 65:362–369. 2010. View Article : Google Scholar : PubMed/NCBI

87 

Spaeth E, Klopp A, Dembinski J, Andreeff M and Marini F: Inflammation and tumor microenvironments: Defining the migratory itinerary of mesenchymal stem cells. Gene Ther. 15:730–738. 2008. View Article : Google Scholar : PubMed/NCBI

88 

Wang J, Ma D, Li Y, Yang Y, Hu X, Zhang W and Fang Q: Targeted delivery of CYP2E1 recombinant adenovirus to malignant melanoma by bone marrow-derived mesenchymal stem cells as vehicles. Anticancer Drugs. 25:303–314. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Jing HX, Duan J, Zhou H, Hu QM and Lei TC: Adipose derived mesenchymal stem cell facilitated TRAIL expression in melanoma treatment in vitro. Mol Med Rep. 14:195–201. 2016.PubMed/NCBI

90 

Seo KW, Lee HW, Oh YI, Ahn JO, Koh YR, Oh SH, Kang SK and Youn HY: Anti-tumor effects of canine adipose tissue-derived mesenchymal stromal cell-based interferon-β gene therapy and cisplatin in a mouse melanoma model. Cytotherapy. 13:944–955. 2011. View Article : Google Scholar : PubMed/NCBI

91 

Tyciakova S, Matuskova M, Bohovic R, Polakova K, Toro L, Skolekova S and Kucerova L: Genetically engineered mesenchymal stromal cells producing TNFα have tumour suppressing effect on human melanoma xenograft. J Gene Med. 17:54–67. 2015. View Article : Google Scholar : PubMed/NCBI

92 

Yi BR, Hwang KA, Aboody KS, Jeung EB, Kim SU and Choi KC: Selective antitumor effect of neural stem cells expressing cytosine deaminase and interferon-beta against ductal breast cancer cells in cellular and xenograft models. Stem Cell Res (Amst). 12:36–48. 2014. View Article : Google Scholar

93 

Yi BR, Kim SU and Choi KC: Additional effects of engineered stem cells expressing a therapeutic gene and interferon-β in a xenograft mouse model of endometrial cancer. Int J Oncol. 47:171–178. 2015.PubMed/NCBI

94 

Yi BR, Park MA, Lee HR, Kang NH, Choi KJ, Kim SU and Choi KC: Suppression of the growth of human colorectal cancer cells by therapeutic stem cells expressing cytosine deaminase and interferon-β via their tumor-tropic effect in cellular and xenograft mouse models. Mol Oncol. 7:543–554. 2013. View Article : Google Scholar : PubMed/NCBI

95 

Kim DJ, Yi BR, Lee HR, Kim SU and Choi KC: Pancreatic tumor mass in a xenograft mouse model is decreased by treatment with therapeutic stem cells following introduction of therapeutic genes. Oncol Rep. 30:1129–1136. 2013.PubMed/NCBI

96 

Yi BR, Kim SU and Choi KC: Co-treatment with therapeutic neural stem cells expressing carboxyl esterase and CPT-11 inhibit growth of primary and metastatic lung cancers in mice. Oncotarget. 5:12835–12848. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2016
Volume 36 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Heo JR, Kim NH, Cho J and Choi KC: Current treatments for advanced melanoma and introduction of a promising novel gene therapy for melanoma (Review). Oncol Rep 36: 1779-1786, 2016
APA
Heo, J., Kim, N., Cho, J., & Choi, K. (2016). Current treatments for advanced melanoma and introduction of a promising novel gene therapy for melanoma (Review). Oncology Reports, 36, 1779-1786. https://doi.org/10.3892/or.2016.5032
MLA
Heo, J., Kim, N., Cho, J., Choi, K."Current treatments for advanced melanoma and introduction of a promising novel gene therapy for melanoma (Review)". Oncology Reports 36.4 (2016): 1779-1786.
Chicago
Heo, J., Kim, N., Cho, J., Choi, K."Current treatments for advanced melanoma and introduction of a promising novel gene therapy for melanoma (Review)". Oncology Reports 36, no. 4 (2016): 1779-1786. https://doi.org/10.3892/or.2016.5032