2,171
Views
25
CrossRef citations to date
0
Altmetric
Commentary

Small molecule drugs with immunomodulatory effects in cancer

&
Pages 2463-2468 | Received 05 May 2015, Accepted 28 May 2015, Published online: 16 Sep 2015

Abstract

Immunotherapy shows promise for positively changing the landscape of the management of many advanced solid tumors, including gastrointestinal (GI) malignancies. Many of these developments have been focused on vaccine-based, monoclonal antibody therapies and more recently, checkpoint inhibitors, although many small molecule inhibitors can function as immunomodulators. Small molecule compounds have several advantages over conventional immunotherapeutic agents including: ease of production and the potential for oral administration. There is a potential niche for small molecule immunomodulators to enhance the efficacy of existing immunotherapeutic and cytotoxic agents. This article focuses on two categories of small molecule compounds with immunomodulatory effects: IDO and MEK inhibitors. Indoleamine -2, 3- dioxygenase (IDO) is known for its effects in tumor immunity. IDO inhibitors are generally well-tolerated and have the potential to enhance anti-tumor responses when combined with checkpoint inhibitors. MEK inhibitors affect signal transduction of the RAS-RAF-MEK pathway and numerous MEK inhibitors are currently being investigated in solid tumors. Small molecule immunomodulators are currently being investigated for their potential role in augmenting the effects of conventional immunotherapeutic agents although further research is required to identify those patients most likely to respond to combination therapy.

Abbreviations

APC=

Antigen presenting cell

BID=

Twice daily

CR=

Complete response

CRC=

Colorectal cancer

CTLA-4=

Cytotoxic T-lymphocyte-associated protein-4

DC=

Dendritic cell

DNA=

DNA

DNMT=

DNA methyltransferase

EGFR=

Epidermal growth factor receptor

FDA=

Food and Drug Administration

GI=

Gastrointestinal

HDAC=

Histone deacetylase complex

IDO=

Indoleamine-2,3-dioxygenase

IFNγ=

Interferon gamma

IV=

Intravenous

LPS=

Lipopolysaccharide

MAPK=

Mitogen-activated protein kinase

1-MT=

1-methyltryptophan

MTD=

Maximum tolerated dose

NSCLC=

Non-small cell lung cancer

PBMC=

Peripheral blood mononuclear cell

PR=

Partial response

RORγt=

RAR-related orphan receptor

TDO=

Tryptophan 2,3-dioxygenase

Th17=

T helper 17 cell

TNFα=

Tumor necrosis factor α

Treg=

Regulatory T cell.

Introduction

Cytotoxic chemotherapies have traditionally been used to treat advanced solid tumors but these are generally non-curative, are associated with significant toxicities and do not produce durable response rates. There is increasing interest in the role of immunotherapy as an attractive strategy in the management of advanced solid tumors previously assumed to be non-immunogenic. Checkpoint inhibitors (ipilimumab, nivolumab) have had unprecedented durable responses in subsets of patients with advanced melanoma and non-small cell lung cancer (NSCLC).Citation1,2 Even patients with metastatic pancreatic cancer, known for its poor survival rates, were recently shown to have improved survival when treated with the GVAX and CRS-207 vaccine therapies.Citation3 While immunotherapy has historically involved cell or antigen-based vaccine therapies and monoclonal antibodies, there has been considerable focus on the development of small molecule drugs which have immunomodulatory effects. Small molecule drugs have the advantage of the potential for oral administration and ease of production relative to standard biologic agents. The preclinical development of small molecule drugs requires extensive knowledge of the biology of a particular target in a given cancer, is amenable to high-throughput screening of thousands of compounds and potentially allows administration of more personalized medicine to patients.Citation4

Currently, there are numerous efforts to develop small molecule inhibitors with immunomodulatory effects in cancer. Many of these compounds are at an early stage of development (). The importance of epigenetic changes in cancer has prompted research efforts into HDAC and DNMT inhibitors. The HDAC inhibitors romidepsin and vorinostat have already been FDA-approved in hematologic malignancies.Citation5 There is a significant interplay between components of the immune system (IFNγ) and the anti-tumor effects of HDAC inhibitors.Citation6 The DNA methyltransferase (DNMT) inhibitors azacitidine and decitabine are FDA-approved in high risk myelodysplastic syndrome.Citation7 Azacitidine has been shown to have immunomodulatory effects in colon cancer by converting CD11b myeloid cells into antigen presenting cells (APCs).Citation8 Thalidomide analogs are small molecule glutamic acid derivatives which have been FDA-approved in multiple myeloma.Citation9 Thalidomide is renowned for its immunomodulatory effects including T cell costimulation and increasing the effects of NK and cytotoxic T cell functions. Toll-like receptor agonists can initiate both innate and adaptive immune systems. Imiquimod is FDA-approved for the treatment of superficial basal cell carcinoma.Citation10 It functions by potently inducing IFNγ and other cytokines leading to activation of APCs and a Th1-driven response.Citation11 RORγt inhibitors function by blocking the function of pro-inflammatory Th17 cells.Citation12 Several small molecule compounds which inhibit RORγt are in development, mostly targeting hematologic malignancies.

Table 1. Categories of small molecule compounds with immunomodulatory functions in solid tumors. Abbreviations: DC: dendritic cell, DNMT: DNA methyltransferase, HDAC: histone deacetylase complex. IDO: indoleamine-2, 3-dioxygenase, RORγt: Retinoic acid receptor-related orphan nuclear receptor gamma, TNFα: tumor necrosis factor α

In this commentary, we will discuss IDO and MEK inhibitors in detail, focusing on gastrointestinal malignancies where appropriate.

IDO Inhibitors

Indoleamine-2, 3-dioxygenase (IDO) is an intracellular enzyme that is involved in the rate-limiting step of the catabolism of L-tryptophan in the kynurenine pathway, which is the first step in the biosynthesis of nicotinamide adenine dinucleotide, an important regulator of metabolism. Its role as an immunomodulator was discovered when it was found to have an important role in preventing allogenic fetal rejection by T cells during pregnancy. Its role in tumor immunity is evidenced by the fact that tumor cells transfected with IDO are not rejected in mice which were immunized with a dominant tumor antigen.Citation13 The expression of IDO is controlled by Bin1, a tumor suppressor gene, and studies show that the loss of Bin1 is associated with increased IDO expression through the STAT1 and NF-κB pathways.Citation14,15 IDO deficiency in a preclinical model of lung cancer is associated with decreased vascularization and immune escape.Citation16

IDO expression has been detected in a variety of cancers including pancreatic and colorectal.Citation17,18 IDO functions by mediating immune escape by suppressing the activation of T cells which are sensitive to tryptophan starvation and kynurenine downstream catabolites.Citation19 While IDO expression occurs in tumors, it also occurs in a subset of plasmacytoid DCs (dendritic cells) in tumor-draining lymph nodes.Citation20

IDO expression in regulatory DCs is prompted by an autocrine interferon process controlled by CTLA-4 pathway receptors on regulatory T cells (T reg). This converts the DC into a more quiescent state and reduces its capacity to present antigens to T cells.Citation21 However, IDO+ DCs are also able to prompt CD4+ T cells to become Tregs. If this occurs in a tumor-draining lymph node, IDO can drive the production of Tregs and reg DCs which will further suppress immunity against tumor cells.

Preclinical studies of 1-MT (1-methyltryptophan), a tryptophan mimetic, showed that it reduced tumor growth but did not prevent tumor progression. However, when combined with cyclophosphamide, there was an additional anti-tumor effect compared to chemotherapy alone.Citation22 Resistance to IDO inhibition can be explained by the use of alternative mechanisms which make up for the loss of IDO expression. Tryptophan-2, 3, - dioxygenase (TDO) is a ubiquitous enzyme with a different structure than IDO but has similar activity in tryptophan metabolism which can also mediate the immune response in tumors.Citation23

Based on preclinical evidence that indoximod, the D isomer of 1-MT, has synergistic effects with chemotherapy in a preclinical model of breast cancer, a phase I study showed that it was well tolerated when combined with docetaxel in 27 patients with pre-treated metastatic solid tumors including pancreatic, rectal and esophageal cancers.Citation24 There were no complete responses, 18% had partial responses, 4% had stable disease >6 months and 36% had progressive disease.

A second IDO inhibitor, INCB024360, is currently the focus of several clinical trials encompassing multiple tumor types (). It is an orally available hydroxyamidine small molecule inhibitor which potently and selectively inhibits IDO1 inhibitor (IC50 = 7.1 nM).Citation25 Preclinical data using pancreatic tumor xenografts showed that INCB024360 reduces tumor growth in immunocompetent but not immunodeficient mice.

Table 2. List of clinical trials of IDO inhibitors in patients with cancer (all trials listed are currently recruiting) MTD: Maximum tolerated dose

The phase I dose-escalation study of INCB024360 (NCT01195311) included 52 patients with multiple tumor types including colorectal (45%) and melanoma (12%).Citation26 Patients received daily doses of INCB024360 with doses ranging from 50 mg once daily to 700 mg BID (twice daily). There was no maximum tolerated dose identified and no objective responses were reported although 15 patients (28%) had stable disease at 56 days. Doses ≥300 mg BID achieved greater than 90% inhibition of IDO1 throughout the dosing period. The most common adverse events were fatigue, nausea, anorexia, vomiting, constipation, abdominal pain, diarrhea, dyspnea, back pain and cough (≥20%). This phase I study also found significant reductions in plasma kynurenine/tryptophan ratios and when whole blood samples collected after treatment were stimulated ex vivo with IFNγ and lipopolysaccharide (LPS), INCB024360 showed >90% inhibition of IDO activity.Citation27 In an effort to detect a biomarker that may guide the selection of IDO inhibitors, a monoclonal antibody (rabbit anti-human) has been generated which can stain human tumor tissue samples and potentially determine which patients are likely to respond to IDO inhibition.Citation28

Correlating with preclinical data which suggested that IDO inhibition is not effective as monotherapy, INCB024360 was combined with the anti-CTLA4 antibody ipilimumab in patients with advanced melanoma in a phase I/II study.Citation29 Preliminary data from this study (NCT01604889) showed that 300 mg BID combined with ipilimumab (3 mg/kg IV every 21 days for 4 cycles) resulted in clinically significant ALT elevations after 30–76 days of treatment but were reversible with steroids and treatment discontinuation. These patients were able to resume therapy albeit with lower doses of INCB024360 (25 mg bid). Six of 8 patients had tumor reductions by the time of first imaging. These preliminary data suggest the role of IDO inhibition in providing enhanced anti-tumor effects with anti-CTLA4 therapy.

Another 2nd generation IDO1 inhibitor, NLG-919, is currently recruiting patients with advanced solid tumors in a phase I clinical trial.Citation30 This drug will be administered orally for 21 days (28 day cycles) with the view to establishing the maximum tolerated dose (MTD).

IDO has also been targeted with peptide vaccine therapy. A phase I study treated 15 HLA-A2-postive patients with metastatic NSCLC with IDO5 vaccines administered every 14 days for 2.5 months and subsequently monthly.Citation31 The vaccine was well tolerated with no grade 3/4 toxicities reported. Vaccinated patients had longer survival than vaccine-untreated patients (25.9 vs. 7.7 months, p = 0.03). Flow cytometry of PBMCs showed significant reduction in the Treg population (p = 0.03) and durable responses were noted with 1 patient developing a partial response after one year of therapy and 6 patients achieving long-lasting disease stability (defined as lasting ≥8.5 months).

IDO has been established as an important player in immune escape in tumor development. It shows limited efficacy when used alone but has significant potential to enhance anti-tumor responses when combined with conventional cytotoxic chemotherapy, peptide-based vaccine therapies and checkpoint inhibitors.

MEK Inhibition

The MAPK (mitogen-activated protein kinase) pathway is one of the most well-defined signaling pathways in human cancer. This pathway consists of kinases which are tightly regulated by a sequence of phosphorylation and activation of protein kinases including the RAS-RAF-MEK1/2- ERK1/2 pathway. MEK1/MEK2 catalyze the phosphorylation of tyrosine and threonine residues in ERK1/ERK2 thereby mediating cellular signals transduced from RAS/RAF.Citation32 Genes encoding RAS (KRAS, NRAS, HRAS) are among the most frequently mutated genes in cancer including biliary tract (33%), large bowel (36%), esophageal (5%), pancreatic (63%) and gastric cancer (12%).Citation33 Dysregulation of the MAPK pathway is associated with effects on the immune system. Animal models of MAPK dysfunction (Mkk3−/−) have evidence of defective IL-12 production by macrophages and dendritic cells.Citation34 The MAPK signaling pathways have also been implicated in the differential regulation of myeloid-derived dendritic cell (MDCC) maturation and the modulation of the Th cells (T helper) toward Th1 or Th2 subsets.Citation35 The phosphorylation of p38 MAPK is essential for the maturation of immature DCs as shown by MAPK inhibitors reducing the DC maturation marker CD83 induced by LPS and TNFα.Citation36

To date, a single MEK inhibitor, trametinib, has been approved by the FDA for its use in unresectable or metastatic melanoma. This was based on a phase III trial in patients with advanced BRAF V600E/V600K melanoma which showed that trametinib improved progression-free and overall survival compared to chemotherapy.Citation37 Numerous other MEK inhibitors have been the subject of clinical trials and have failed to substantially improve survival outcomes in multiple tumor types. Similar to IDO inhibitors, MEK inhibitors have limited efficacy as monotherapy and require combination with other targeted therapies/chemotherapy to achieve their optimal efficacy. The degree of MEK1/2 inhibition to achieve a clinical response is also not known.

A number of MEK inhibitors are the subject of ongoing clinical trials (). Trametinib is a second generation MEK inhibitor shown to potently inhibit p-ERK 1/2 in vitro. The combination of trametinib and dabrafenib has been shown to improve overall survival in metastatic melanoma compared to vemurafenib alone.Citation38 A phase III study is ongoing combining trametinib, dabrafenib (BRAF inhibitor) and panitumumab (anti-EGFR) in patients with BRAF V600E mutant colorectal cancer (CRC). Preliminary data have shown that this combination is tolerable and clinical activity (partial response (PR) /stable disease) has been reported.Citation39

Table 3. Selection of clinical trials of MEK inhibitors in patients with solid tumors (all trials listed are currently recruiting)

Selumetinib is a potent, selective ATP non-competitive MEK inhibitor. It was combined with irinotecan in the second line setting in 31 patients with KRAS mutant metastatic CRC and was shown to have clinical activity (9.7% had partial response, 51.6% had disease stability).Citation40 Despite strong preclinical data of combination MEK and Akt inhibition, the combination of selumetinib and MK-2206 (Akt 1/2/3 inhibitor) did not result in clinical activity in patients with metastatic CRC.Citation41 Selumetinib was shown to be tolerable in a phase II study of 28 patients with advanced biliary cancers and 3 patients (12%) had objective responses with 14 patients having prolonged stable disease (>16 weeks).Citation42

MEK162 (Binimetinib), a selective ATP-noncompetitive MEK1/2 inhibitor, is the subject on numerous ongoing clinical trials (). It is similar to other MEK inhibitors in terms of its toxicity. A phase I study in 28 patients with unresectable/locally advanced or metastatic biliary cancer received MEK162 and clinical responses were seen (1 CR, 1 PR, 11 stable disease).Citation43 This strategy has progressed to a phase I/II study in advanced biliary cancer combining MEK162 with a standard chemotherapy combinations gemcitabine/cisplatin (NCT01828034) and gemcitabine/oxaliplatin (NCT02105350). MEK162 is also the subject of ongoing trials in metastatic CRC in combination with FOLFOX chemotherapy (NCT02041481) and panitumumab (NCT01927341).

Pimasertib is a selective non-competitive inhibitor of MEK 1/2. It had shown promising activity in preclinical studies of pancreatic cancer but a phase II placebo-controlled randomized study of gemcitabine with pimasertib vs. placebo did not show an improvement in PFS.Citation44 Refametinib is a highly selective allosteric MEK1/2 inhibitor which has shown anti-tumor activity in preclinical studies involving numerous tumor types. A phase II study combining refametinib with gemcitabine showed a trend toward clinical benefit in patients with wild-type KRAS metastatic pancreatic cancer.Citation45 Additional MEK inhibitors which have been mostly studied in non-GI cancers to date include: PD0325901, cobimetinib (studied mostly in melanoma), R04987655, AZD08330, TAK-733 and GDC-0623.

In general, MEK inhibitors are well tolerated although several toxicities can occur in common with other kinase inhibitors e.g. rash, diarrhea, fatigue.Citation46 There have been reports of specific ocular toxicities associated with MEK inhibitors which can present with visual loss/blurred vision. Of particular concern are optic neuropathy, retinal venous occlusion and central serous retinopathy which require urgent specialist referral.Citation47 Serious, but rarer, toxicities include dropped-head syndrome,Citation48 markedly elevated CPK levelsCitation49 and left ventricular dysfunction.Citation50

Conclusion

The management of selected advanced solid tumors has been revolutionized by immunotherapy. To date, this has been mostly compromised of vaccine-based therapies and monoclonal antibodies. There is a role in this niche for small molecule compounds to augment these effects on the immune system either alone or in combination with cytotoxic chemotherapy/other immune therapies. Numerous compounds are currently in development and many have reached phase III clinical trials or have been approved by the FDA. However, the lack of a biomarker to predict responsiveness to these therapies, hinders their development and collaborative research investigating potential biomarkers is warranted. Future directions may include the development of small molecule checkpoint inhibitors. Given the advantages that small molecule drugs possess in terms of their relative ease of administration, further research and development of small molecules with immunomodulatory effects is worthwhile in order to improve options for patients with advanced solid tumors.

Disclosure of Potential Conflicts of Interest

Under a licensing agreement between Aduro Biotech and the Johns Hopkins University (University), the University and investigators are entitled to milestone payments and royalty on sales of the GM-CSF-secreting tumor vaccine products (GVAX) described herein. Dr. Zheng received a research grant from iTeos Therapeutics.

Funding

This work was supported in part by the NIH K23 CA148964-01 (L.Z.), NIH 1 R01 CA197296-01 (L.Z.), Viragh Foundation and the Skip Viragh Pancreatic Cancer Center at Johns Hopkins (L.Z.), the NCI SPORE in Gastrointestinal Cancers P50 CA062924 (L.Z.).

References

  • 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. New Engl J Med 2011; 364:2517-26; PMID:21639810; http://dx.doi.org/10.1056/NEJMoa1104621
  • Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. New Engl J Med 2012; 366:2455-65; PMID:22658128; http://dx.doi.org/10.1056/NEJMoa1200694
  • Le DT, Wang-Gillam A, Picozzi V, Greten TF, Crocenzi T, Springett G, Morse M, Zeh H, Cohen D, Fine RL, et al. Safety and survival with GVAX pancreas prime and listeria monocytogenes-expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J Clin Oncol 2015; 33:1325-33; PMID:25584002; http://dx.doi.org/10.1200/JCO.2014.57.4244
  • Hoelder S, Clarke PA, Workman P. Discovery of small molecule cancer drugs: successes, challenges and opportunities. Mol Oncol 2012; 6:155-76; PMID:22440008; http://dx.doi.org/10.1016/j.molonc.2012.02.004
  • Whittaker SJ, Demierre MF, Kim EJ, Rook AH, Lerner A, Duvic M, Scarisbrick J, Reddy S, Robak T, Becker JC, et al. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol 2010; 28:4485-91; PMID:20697094; http://dx.doi.org/10.1200/JCO.2010.28.9066
  • West AC, Mattarollo SR, Shortt J, Cluse LA, Christiansen AJ, Smyth MJ, Johnstone RW. An intact immune system is required for the anticancer activities of histone deacetylase inhibitors. Cancer Res 2013; 73:7265-76; PMID:24158093; http://dx.doi.org/10.1158/0008-5472.CAN-13-0890
  • Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A, Schoch R, Gattermann N, Sanz G, List A, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol 2009; 10:223-32; PMID:19230772; http://dx.doi.org/10.1016/S1470-2045(09)70003-8
  • Daurkin I, Eruslanov E, Vieweg J, Kusmartsev S. Generation of antigen-presenting cells from tumor-infiltrated CD11b myeloid cells with DNA demethylating agent 5-aza-2'-deoxycytidine. Cancer Immunol Immunotherapy 2010; 59:697-706; PMID:19882154; http://dx.doi.org/10.1007/s00262-009-0786-4
  • Rajkumar SV, Blood E, Vesole D, Fonseca R, Greipp PR, Eastern cooperative oncology G. phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the eastern cooperative oncology group. J Clin Oncol 2006; 24:431-6; PMID:16365178; http://dx.doi.org/10.1200/JCO.2005.03.0221
  • Eigentler TK, Kamin A, Weide BM, Breuninger H, Caroli UM, Mohrle M, Radny P, Garbe C. A phase III, randomized, open label study to evaluate the safety and efficacy of imiquimod 5% cream applied thrice weekly for 8 and 12 weeks in the treatment of low-risk nodular basal cell carcinoma. J Am Acad Dermatol 2007; 57:616-21; PMID:17610993; http://dx.doi.org/10.1016/j.jaad.2007.05.022
  • Schon MP, Schon M. Imiquimod: mode of action. British J Dermatol 2007; 157 2:8-13; http://dx.doi.org/10.1111/j.1365-2133.2007.08265.x
  • Huh JR, Littman DR. Small molecule inhibitors of RORgammat: targeting Th17 cells and other applications. Europ J Immunol 2012; 42:2232-7; PMID:22949321; http://dx.doi.org/10.1002/eji.201242740
  • Uyttenhove C, Pilotte L, Theate I, Stroobant V, Colau D, Parmentier N, Boon T, Van den Eynde BJ. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med 2003; 9:1269-74; PMID:14502282; http://dx.doi.org/10.1038/nm934
  • Muller AJ, DuHadaway JB, Donover PS, Sutanto-Ward E, Prendergast GC. Inhibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat Med 2005; 11:312-9; PMID:15711557; http://dx.doi.org/10.1038/nm1196
  • Prendergast GC. Immune escape as a fundamental trait of cancer: focus on IDO. Oncogene 2008; 27:3889-900; PMID:18317452; http://dx.doi.org/10.1038/onc.2008.35
  • Smith C, Chang MY, Parker KH, Beury DW, DuHadaway JB, Flick HE, Boulden J, Sutanto-Ward E, Soler AP, Laury-Kleintop LD, et al. IDO is a nodal pathogenic driver of lung cancer and metastasis development. Cancer Discov 2012; 2:722-35; PMID:22822050; http://dx.doi.org/10.1158/2159-8290.CD-12-0014
  • Witkiewicz A, Williams TK, Cozzitorto J, Durkan B, Showalter SL, Yeo CJ, Brody JR. Expression of indoleamine 2,3-dioxygenase in metastatic pancreatic ductal adenocarcinoma recruits regulatory T cells to avoid immune detection. J Am College Surgeons 2008; 206:849-54; discussion 54-6; PMID:18471709; http://dx.doi.org/10.1016/j.jamcollsurg.2007.12.014
  • Ferdinande L, Decaestecker C, Verset L, Mathieu A, Moles Lopez X, Negulescu AM, Van Maerken T, Salmon I, Cuvelier CA, Demetter P. Clinicopathological significance of indoleamine 2,3-dioxygenase 1 expression in colorectal cancer. British J Cancer 2012; 106:141-7; PMID:22108515; http://dx.doi.org/10.1038/bjc.2011.513
  • Fallarino F, Grohmann U, Vacca C, Bianchi R, Orabona C, Spreca A, Fioretti MC, Puccetti P. T cell apoptosis by tryptophan catabolism. Cell Death Differentiation 2002; 9:1069-77; PMID:12232795; http://dx.doi.org/10.1038/sj.cdd.4401073
  • Munn DH, Sharma MD, Hou D, Baban B, Lee JR, Antonia SJ, Messina JL, Chandler P, Koni PA, Mellor AL. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest 2004; 114:280-90; PMID:15254595; http://dx.doi.org/10.1172/JCI200421583
  • Sharma MD, Baban B, Chandler P, Hou DY, Singh N, Yagita H, Azuma M, Blazar BR, Mellor AL, Munn DH. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J Clinical Investigation 2007; 117:2570-82; PMID:17710230; http://dx.doi.org/10.1172/JCI31911
  • Hou DY, Muller AJ, Sharma MD, DuHadaway J, Banerjee T, Johnson M, Mellor AL, Prendergast GC, Munn DH. Inhibition of indoleamine 2,3-dioxygenase in dendritic cells by stereoisomers of 1-methyl-tryptophan correlates with antitumor responses. Cancer Res 2007; 67:792-801; PMID:17234791; http://dx.doi.org/10.1158/0008-5472.CAN-06-2925
  • Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, Schumacher T, Jestaedt L, Schrenk D, Weller M, et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 2011; 478:197-203; PMID:21976023; http://dx.doi.org/10.1038/nature10491
  • Soliman HH, Jackson E, Neuger T, Dees EC, Harvey RD, Han H, Ismail-Khan R, Minton S, Vahanian NN, Link C, et al. A first in man phase I trial of the oral immunomodulator, indoximod, combined with docetaxel in patients with metastatic solid tumors. Oncotarget 2014; 5:8136-46; PMID:25327557
  • Liu X, Shin N, Koblish HK, Yang G, Wang Q, Wang K, Leffet L, Hansbury MJ, Thomas B, Rupar M, et al. Selective inhibition of IDO1 effectively regulates mediators of antitumor immunity. Blood 2010; 115:3520-30; PMID:20197554; http://dx.doi.org/10.1182/blood-2009-09-246124
  • Beatty GL, O'Dwyer PJ, Clark J, Shi JG, Newton RC, Schaub R, Maleski J, Leopold L, Gajewski T. Phase I study of the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of the oral inhibitor of indoleamine 2,3-dioxygenase (IDO1) INCB024360 in patients (pts) with advanced malignancies. J Clin Oncol 2013; 31 (suppl; abstr 3025).
  • Newton RC, Scherle PA, Bowman K, Liu X, Beatty GL, O'Dwyer PJ, Gajewski T, Bowman J, Schaub R, Leopold L. Pharmacodynamic assessment of INCB024360, an inhibitor of indoleamine 2,3-dioxygenase 1 (IDO1), in advanced cancer patients. J Clin Oncol 30, 2012 (suppl; abstr 2500^)
  • Hiscox AG H, Couto J, Liao Z, Zhu Y, Liu X, Newton RC, Hnatyszyn J. Development of an IHC-based detection method for studying indoleamine 2,3-dioxygenase 1 (IDO1) expression in human cancers. 2014 ASCO annual meeting. J Clin Oncol 2014:5s 3043
  • Gibney GTHO, Gangadhar TC, Lutzky J, Olszanski AJ, Gajewski T, Chmielowski B, Boasberg PD, Zhao Y, Newton RC, Scherle PA, et al. Preliminary results from a phase 1/2 study of INCB024360 combined with ipilimumab (ipi) in patients (pts) with melanoma. J Clin Oncol 2014; 32:3010
  • Khleif SMD, Nyak-Kapoor A, Mautino MR, Kennedy E, Vahanian NN, Link CJ. First-in-human phase 1 study of the novel indoleamine-2,3-dioxygenase (IDO) inhibitor NLG-919. J Clin Oncol 2014; 32:5s:TPS3121
  • Iversen TZ, Engell-Noerregaard L, Ellebaek E, Andersen R, Larsen SK, Bjoern J, Zeyher C, Gouttefangeas C, Thomsen BM, Holm B, et al. Long-lasting disease stabilization in the absence of toxicity in metastatic lung cancer patients vaccinated with an epitope derived from indoleamine 2,3 dioxygenase. Clin Cancer Res 2014; 20:221; PMID:24218513; http://dx.doi.org/10.1158/1078-0432.CCR-13-1560
  • Roskoski R Jr. ERK1/2 MAP kinases: structure, function, and regulation. Pharmacol Res 2012; 66:105-43; PMID:22569528; http://dx.doi.org/10.1016/j.phrs.2012.04.005
  • Prior IA, Lewis PD, Mattos C. A comprehensive survey of Ras mutations in cancer. Cancer Res 2012; 72:2457-67; PMID:22589270; http://dx.doi.org/10.1158/0008-5472.CAN-11-2612
  • Engel MA, Neurath MF. Anticancer properties of the IL-12 family–focus on colorectal cancer. Current Med Chem 2010; 17:3303-8; PMID:20712574; http://dx.doi.org/10.2174/092986710793176366
  • Puig-Kroger A, Relloso M, Fernandez-Capetillo O, Zubiaga A, Silva A, Bernabeu C, Corbi AL. Extracellular signal-regulated protein kinase signaling pathway negatively regulates the phenotypic and functional maturation of monocyte-derived human dendritic cells. Blood 2001; 98:2175-82; PMID:11568005; http://dx.doi.org/10.1182/blood.V98.7.2175
  • Arrighi JF, Rebsamen M, Rousset F, Kindler V, Hauser C. A critical role for p38 mitogen-activated protein kinase in the maturation of human blood-derived dendritic cells induced by lipopolysaccharide, TNF-α, and contact sensitizers. J Immunol 2001; 166:3837-45; PMID:11238627; http://dx.doi.org/10.4049/jimmunol.166.6.3837
  • Flaherty KT, Robert C, Hersey P, Nathan P, Garbe C, Milhem M, Demidov LV, Hassel JC, Rutkowski P, Mohr P, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. New Engl J Med 2012; 367:107-14; PMID:22663011; http://dx.doi.org/10.1056/NEJMoa1203421
  • Robert C, Karaszewska B, Schachter J, Rutkowski P, Mackiewicz A, Stroiakovski D, Lichinitser M, Dummer R, Grange F, Mortier L, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. New Engl J Med 2015; 372:30-9; PMID:25399551; http://dx.doi.org/10.1056/NEJMoa1412690
  • Bendell JCACE, André T, Tabernero J, Gordon MS, Bernards R, Van Cutsem E, Tejpar S, Sidhu R, Go WY, Allred A, et al. Efficacy and tolerability in an open-label phase I/II study of MEK inhibitor trametinib (T), BRAF inhibitor dabrafenib (D), and anti-EGFR antibody panitumumab (P) in combination in patients (pts) with BRAF V600E mutated colorectal cancer (CRC). J Clin Oncol 32:5s 2014 3515
  • Hochster HS, Uboha N, Messersmith W, Gold PJ, BH ON, Cohen D, Denlinger C, Cohen S, Leichman CG, Leichman L, et al. Phase II study of selumetinib (AZD6244, ARRY-142886) plus irinotecan as second-line therapy in patients with K-RAS mutated colorectal cancer. Cancer Chemotherapy Pharmacol 2015; 75:17-23; http://dx.doi.org/10.1007/s00280-014-2609-3
  • Do K, Speranza G, Bishop R, Khin S, Rubinstein L, Kinders RJ, Datiles M, Eugeni M, Lam MH, Doyle LA, et al. Biomarker-driven phase 2 study of MK-2206 and selumetinib (AZD6244, ARRY-142886) in patients with colorectal cancer. Investigational New Drugs 2015; 33(3): 720–8; PMID:25637165
  • Bekaii-Saab T, Phelps MA, Li X, Saji M, Goff L, Kauh JS, O'Neil BH, Balsom S, Balint C, Liersemann R, et al. Multi-institutional phase II study of selumetinib in patients with metastatic biliary cancers. J Clin Oncol 2011; 29:2357-63; PMID:21519026; http://dx.doi.org/10.1200/JCO.2010.33.9473
  • Finn RSJMM, Tan, BR, Weekes CD, Bendell JC; Patnaik A, Khan GN, Laheru D, Anderson L, Christy-Bittel JL, Barrett E et al. A phase I study of MEK inhibitor MEK162 (ARRY-438162) in patients with biliary tract cancer. 2010 ASCO GI conference. J Clin Oncol 2012:4 220
  • Van Cutsem EH, Bazin MI, Canon JL, Poddubskaya E, Manojlovic N, Milella M, Radenkovic D, Verslype C, Guo W, Damstrup L, et al. Phase II randomized trial of MEK inhibitor pimasertib or placebo combined with gemcitabine in the first-line treatment of metastatic pancreatic cancer. J Clin Oncol 2015; 33:3 344
  • Van Laethem JL, Jassem J, Heinemann V, Weekes CD, Bridgewater JA, Cascinu S, Melichar B, Peeters M, Ross PJ, Saramak P, Giurescu M, et al. Phase II study of refametinib (BAY 86-9766), an allosteric dual MEK 1/2 inhibitor, and gemcitabine in patients with unresectable, locally advanced, or metastatic pancreatic cancer. J Clin Oncol 2014; 32 (suppl; abstr 4025).
  • Wang D, Boerner SA, Winkler JD, LoRusso PM. Clinical experience of MEK inhibitors in cancer therapy. Biochimica et Biophysica Acta 2007; 1773:1248-55; PMID:17194493; http://dx.doi.org/10.1016/j.bbamcr.2006.11.009
  • Renouf DJ, Velazquez-Martin JP, Simpson R, Siu LL, Bedard PL. Ocular toxicity of targeted therapies. J Clin Oncol 2012; 30:3277-86; PMID:22649132; http://dx.doi.org/10.1200/JCO.2011.41.5851
  • Chen X, Schwartz GK, DeAngelis LM, Kaley T, Carvajal RD. Dropped head syndrome: report of three cases during treatment with a MEK inhibitor. Neurology 2012; 79:1929-31; PMID:23077008; http://dx.doi.org/10.1212/WNL.0b013e318271f87e
  • Ascierto PA, Schadendorf D, Berking C, Agarwala SS, van Herpen CM, Queirolo P, Blank CU, Hauschild A, Beck JT, St-Pierre A, et al. MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 study. Lancet Oncol 2013; 14:249-56; PMID:23414587; http://dx.doi.org/10.1016/S1470-2045(13)70024-X
  • Chen MH, Kerkela R, Force T. Mechanisms of cardiac dysfunction associated with tyrosine kinase inhibitor cancer therapeutics. Circulation 2008; 118:84-95; PMID:18591451; http://dx.doi.org/10.1161/CIRCULATIONAHA.108.776831

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.