Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

Review

1.

Introduction

2.

Current experience with PDGFR inhibitors

3.

Clinical development of new PDGFR targeted molecules

4.

Conclusion

5.

Expert opinion

Investigational therapies up to Phase II which target PDGF receptors: potential anti-cancer therapeutics Jennifer Arrondeau, Olivier Huillard, Camille Tlemsani, Anatole Cessot, Pascaline Boudou-Rouquette, Benoit Blanchet, Audrey Thomas-Schoemann, Michel Vidal, Jean-Marie Tigaud, Jean-Philippe Durand, Jerome Alexandre & Francois Goldwasser† †

Paris Descartes University, Cochin Hospital, AP-HP, Medical Oncology Department, Angiogenesis Inhibitors Multidisciplinary Study Group (CERIA), Paris, France

Introduction: The platelet-derived growth factor receptor (PDGFR) pathway has important functions in cell growth and, by overexpression or mutation, could also be a driver for tumor development. Moreover, PDGFR is expressed in a tumoral microenvironment and could promote tumorigenesis. With these biological considerations, the PDGFR pathway could be an interesting target for therapeutics. Currently, there are many molecules under development that target the PDGFR pathway in different types of cancer. Areas covered: In this review, the authors report the different molecules under development, as well as those approved albeit briefly, which inhibit the PDGFR pathway. Furthermore, the authors summarize their specificities, their toxicities, and their development. Expert opinion: Currently, most PDGFR kinase inhibitors are multikinase inhibitors and therefore do not simply target the PDGFR pathway. The development of more specific PDGFR inhibitors could improve drug efficacy. Moreover, selecting tumors harboring mutations or amplifications of PDGFR could improve outcomes associated with the use of these molecules. The authors believe that new technologies, such as kinome arrays or pharmacologic assays, could be of benefit to understanding resistance mechanisms and develop more selective PDGFR inhibitors. Keywords: anti-cancer therapies, platelet-derived growth factor, platelet-derived growth factor receptor pathway, targeted therapies Expert Opin. Investig. Drugs [Early Online]

1.

Introduction

The deregulated protein tyrosine kinase activity is of major importance in the pathogenesis of human cancers. Targeted therapy has transformed the approach of the management of various cancers and represents a therapeutic breakthrough. Among tyrosine kinases, the platelet-derived growth factor (PDGF) and its receptors (platelet-derived growth factor receptor [PDGFR]) play critical roles in the regulation of cancer cell processes and are increasingly the target of investigational therapies. This review focuses on the development of PDGFR inhibitors with emphasis on future directions that may help such development. The PDGFR pathway The PDGF family has different ligands, consisting of one of four polypeptide chains: PDGF-A, PDGF-B, PDGF-C and PDGF-D [1]. Each chain is encoded by 1.1

10.1517/13543784.2015.1005736 © 2015 Informa UK, Ltd. ISSN 1354-3784, e-ISSN 1744-7658 All rights reserved: reproduction in whole or in part not permitted

1

J. Arrondeau et al.

Article highlights. .

.

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

.

The platelet-derived growth factor receptor pathway could be a driver for tumor development, by overexpression or mutation or by expression in tumoral microenvironment. Currently, most of PDGFR kinase inhibitors under development are multikinase inhibitors. Selecting tumors harboring mutations or amplifications of PDGFR could improve outcomes associated with the use of these molecules.

This box summarizes key points contained in the article.

an individual gene located on chromosomes 7, 22, 4, and 11 respectively [2]. These polypeptide chains form homo- or heterodimers: PDGF-AA, PDGF-AB, PDGF-BB, PDGFCC, and PDGF-DD. These PDGF isoforms exert their cellular effects by binding to PDGF-a and PDGF-b protein kinase receptors (PDGFRa and PDGFRb). Dimerization of receptors occurs after ligand binding, which can be homo- or heterodimerization (PDGFR-aa, PDGFR-ab, or PDGFR-bb). The PDGF polypeptide chains binds to the receptors with different affinities; PDGF-AA, -BB, -AB and --CC induce aa homodimer receptor, PDGF-BB and --DD induce bb homodimer receptor, and PDGF-AB, -BB, -CC and --DD induce ab homodimer receptor [3]. The interaction of the ligand and its receptor leads to the autophosphorylation of the tyrosine kinase domain activating the intracellular PDGF pathway (Figure 1). When phosphorylated, the tyrosine kinase domain recruits the SH-2 domain containing signal transduction proteins and activates signaling molecules with intrinsic enzymatic activities, such as tyrosine kinase of the Src family, phosphatitdylinositol 3¢-kinase, phospholipase C g 1 and the GTPase activating protein for RAS. The activation of these signaling pathways leads to cell proliferation, survival and cell migration. Therefore, PDGF signaling is involved in many processes in embryonic development and studies in mice with PDGF isoforms or knocked-out receptors have elucidated important roles for this pathway. Evidence for targeting the PDGF pathway for the treatment of cancer and preclinical data

1.2

Overactivity of PDGF signaling can result from mutations or overexpression of PDGF or PDGFR and can lead to tumorigenesis in three manners: autocrine signaling, tumor angiogenesis and elevation of interstitial fluid pressure in tumors (Figure 2). In vitro, selective inhibition of PDGF can inhibit angiogenesis in a sponge angiogenesis model [4]. In vivo, it has been shown that PDGF can stimulate angiogenesis [5] and promote the recruitment of tumor pericytes in knockout mice models [6]. Moreover PDGF has a role in the control of tissue interstitial fluid, and activation of the PDGFR leads to the elevation of interstitial fluid pressure in tumors. 2

The consequence of this elevation is mainly pharmacological and could decrease drug delivery to tumors. Importantly, it has been described that during epithelial-mesenchymal transition, tumor expression of PDGFR increases, leading to the conclusion that epithelial tumors that initially did not respond to PDGF stimulation may be reversible and become responsive to PDGF stimulation [7]. PDGFR activation is documented across many tumor types and may result from mutation, gene fusion, or overexpression. This has mainly been described in the following tumor types: gastrointestinal stromal tumors (GIST) [8], gliomas [9], dermatofibrosarcoma protuberance (DFSP) [10], others types of sarcomas (soft tissue sarcoma, osteosarcoma, synovial sarcoma, Ewing’s sarcoma), hematological diseases [11], prostate cancer [12], liver cancer [13], non-small cell lung cancer [14], breast cancer [15], ovarian cancer [16], and colorectal cancer [17]. Furthermore, stroma cells of solid tumors, including cancer-associated fibroblasts (CAFs), could contribute to tumorigenesis [18]. PDGFR are expressed on CAFs and several reports have described that PDGF stimulation could affect CAF functions. Among these reports, one has shown that tumor-cell derived PDGF recruited CAFs in xenograft studies of breast [19] and lung carcinomas [20]. It has also been reported that transgenic PDGF expression in mouse liver cells resulted in tumor fibrosis and promoted development of hepatocellular carcinoma [21]. Moreover, in mouse models, expression of PDGF promoted recruitment of CAFs and growth of malignant melanoma [22] and liver metastasis of colorectal cancer [23]. All these observations have led to the development of different inhibitors of PDGF signaling and preclinical studies have been performed across different cancer types. Most of these agents, particularly molecules currently approved, are multikinases inhibitors without selective targeting of PDGFR. In glioblastoma cell lines or xenograft models, PDGFR tyrosine kinase inhibitors induced reduction of tumor growth [24]. In prostate cancer, human prostate cancer cells (PC-3) in nude mice showed expression of activated PDGFR and PDGF; treatment with imatinib has led to lower tumor incidence, to smaller tumors and to fewer lymph node metastases compared with control mice [25]. In a mouse model of human ovarian cancer, SU6668, a multikinase inhibitor including PDGFR, exhibited anti-tumor effects, with reduction in tumor growth, ascites production, and metastatic spread [26]; improvement in overall survival (OS) has been demonstrated with SU6668 in mouse models of peritoneally disseminated ovarian cancer [27]. In DFSP tumors, imatinib treatment in cell cultures derived from human DFSP tumors has shown growth reduction [28]. Recently, a more potent selective inhibitor of PDGFR, CP-673451, was effective in vitro to suppress cell viability, to induce cell apoptosis, and to inhibit cell migration in non-small cell lung carcinoma cells and invasion at suppressing non-small cell lung carcinoma tumor growth in vivo [29].

Expert Opin. Investig. Drugs (2015) 24(5)

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

Investigational therapies up to Phase II which target PDGF receptors: potential anti-cancer therapeutics

PDGF-AA

PDGF-AB

PDGF-CC

PDGF-BB

PDGF-AB

PDGF-CC

PDGF-BB

PDGF-BB

PDGF-DD

PDGF-DD

α

α

Src

α

PI3K

β

β

PL-Cγ

β

RAS

- Cell proliferation - Survival - Migration

Figure 1. PDGF pathway with PDGF receptors and its ligands. PDGF: Platelet-derived growth factor; PI3K: Phosphatitdylinositol 3¢-kinase; PL-Cg: Phospholipase C g.

2.

Current experience with PDGFR inhibitors

Currently approved PDGFR inhibitors are multikinase inhibitors also targeting mostly VEGFR or KIT (Figure 2). These therapies are used in different cancer types and can be under development for others. Imatinib is currently approved for the treatment of GIST (in the adjuvant and metastatic settings) and DFSP. It targets VEGFR, PDGFR and KIT. It was first developed for the treatment of chronic myelogenous leukemia (CML) where its efficacy is due to BCR-ABL targeting. It was then developed in GIST for its ability to inhibit cKIT and PDGFR-a as 5% of GISTs have PDGFR-a mutations, which are exclusive with KIT mutations. Different mutations of PDGFR-a have been described, displaying more or less responsiveness to imatinib. A pooled analysis of 289 PDGFR-a mutant GIST patients suggested that more than one-third of these patients may respond to imatinib [30]. Dose limiting toxicities consisted in vomiting, nausea, edema, or dyspnea. Interestingly, imatinib has been under development in prostate cancer and a Phase II trial evaluated the association of imatinib and docetaxel for castration-resistant prostate cancer with bone metastases. The trial was stopped because of excessive toxicity. In this study, phosphorylated PDGFR (p-PDGFR) was measured in peripheral blood leukocytes. Surprisingly, the expected increased probability of p-PDGFR decline in the imatinib group was associated with lower probability of PSA decline,

shorter progression-free survival and OS. In the control arm treated with docetaxel alone, p-PDGFR expression increased, which correlated with improvement of progression-free survival and OS [31]. These observations led to the hypothesis that imatinib-induced p-PDGFR inhibition could antagonize docetaxel efficacy. In ovarian cancer, several Phase II trials have been performed, but most have shown no or minimal efficacy [32]. Imatinib is currently under development in Phase II trials in non-small cell lung carcinoma, mesothelioma, renal cell carcinoma, and head and neck cancer. Other tyrosine kinase inhibitors targeting PDGFR are approved in different cancer types such as axitinib approved in advanced renal cell carcinoma, pazopanib approved in renal cell carcinoma and in soft tissue sarcoma, regorafenib approved in metastatic colorectal cancer and GIST, sorafenib approved in renal cell carcinoma, hepatocellular carcinoma and differentiated thyroid neoplasm, and sunitinib approved in GIST, renal cell carcinoma and pancreatic neuroendocrine tumors. Because the tyrosine kinases targeted by these therapies are very similar, the toxicity profiles are also similar with mainly hypertension, diarrhea, fatigue, nausea, or hand--foot syndrome. As described above, currently approved therapies targeting PDGFR are not selectively targeted on PDGFR, and except for PDGFR mutated GIST and DFSP, the efficacy that has been demonstrated in clinical trials may mostly rely on the inhibition of other tyrosine kinases as VEGFR.

Expert Opin. Investig. Drugs (2015) 24(5)

3

J. Arrondeau et al.

Olaratumab Tovetumab

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

Activating mutation amplification translocation

α

α

β

Axitinib Crenolanib Dastinib Dovitinib Lenvatinib Masatinib Motesanib

Imatinib

Nilotinib Nitedanib Pazopabib Ponatinib Sorafenib Sunitinib Tivozanib

β

Linifanib Orantinib Regorafenib Telatinib Vatalanib

Src

PI3K

PL-Cγ

RAS

STAT

AKT

PKC

RAF MEK

mTOR

ERK

Figure 2. Abnormal PDGF receptors and mechanism of action of PDGFR inhibitors. PDGF: Platelet-derived growth factor; PDGFR: PDGF receptor; PI3K: Phosphatitdylinositol 3¢-kinase; PKC: Protein Kinase C; PL-Cg: Phospholipase C g.

Clinical development of new PDGFR targeted molecules 3.

All trials evaluating PDGFR inhibitor were found on the ClinicalTrials website (ClinicalTrials.gov). Several tyrosine kinase inhibitors and two monoclonal antibodies are under development as shown in Table 1 and their mechanism of action is described in Figure 2. For each molecule, molecular targets and IC50 measure are presented in Table 2 [33-50]. Finally, as shown in Table 3, few molecules are currently studied in Phase II trials for their specific targeting of the PDGFR pathway. The main adverse events reported in the articles of these molecules under development are summarized in Table 4. Phase I and II trials of tyrosine kinase inhibitors which target PDGFR

3.1

Dasatinib is currently approved in hematologic disease (CML and acute lymphocytic leukemia) and is evaluated for the 4

treatment of different solid tumor types. Used as monotherapy, Phase II trials have shown limited activity of dasatinib in several tumor types [51,52] [53-57] and toxicity appears to be important. Current development is mainly focused on combination with chemotherapy or targeted agents in Phase I and II trials. A Phase III trial has evaluated dasatinib in combination with docetaxel in chemotherapy-naive metastatic castration-resistant prostate cancer patients, but showed no survival benefit [58]. Nilotinib is approved for the treatment of chronic myelogenous leukemia. A preclinical study has tested nilotinib in ovarian cancer cells and showed significant inhibition of cell growth in PDGFR-a-positive ovarian cancer cell lines [59]. It has been developed in GIST with Phase II and III trials, with no clinical efficacy demonstrated [60]. Phase II trials are ongoing in melanoma and glioma but only with PDGFR amplification. Nilotinib is generally well tolerated. No combination trials in solid tumors are currently ongoing. Ponatinib is approved for the treatment of CML and acute lymphocytic leukemia. Several Phase II studies are ongoing in different cancer types and especially in GIST with PDGFR

Expert Opin. Investig. Drugs (2015) 24(5)

Investigational therapies up to Phase II which target PDGF receptors: potential anti-cancer therapeutics

Table 1. PDGFR inhibitors currently under development. Molecule

Phase I trials

Crenolanib

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

Dasatinib

NSCLC Pancreas GIST

Dovitinib

Lenvatinib

Linifanib

NSCLC Melanoma

NSCLC

Masitinib

Motesanib

Gallbladder Breast

Nilotinib

Nindetanib

NSCLC HCC Glioblastoma

Phase II trials GliomasO GISTO NSCLCO NSCLCR PancreasR,O ProstateR,O OvarianR,O BreastR,O Head and neckR GlioblastomaR MelanomaO EndometrialO GISTR BreastO EndometrialO BladderO ProstateO Salivary glandsO Adrenocortical carcinomaO CRCO StomachO HCCO Biliary tractO NSCLCO MesotheliomaO GlioblastomaO OvarianO MelanomaO HCCO RCCO NSCLCO GliomaO EndometrialO CRCR NSCLCR,O HCCR BreastO PancreasR OvarianR GISTR ThyroidR MelanomaO GlioblastomaO Giant cell tumorO ProstateR GliomasR,O BreastR,O OvarianR,O CRCR,O ThyroidO CervixO Lung (S/NSCLC) O

Table 1. PDGFR inhibitors currently under development (continued). Phase III trials

Molecule

Phase I trials

Ponatinib Prostate

Olaratumab

Orantinib Telatinib Tivozanib

HCCO O

Breast

Tovetumab Vatalanib

ThyroidR HCCO

Phase III trials

MesotheliomaO EsophagogastricO NSCLCO GISTO ThyroidO GISTR GlioblastomaR NSCLCO OvarianO Soft tissueO SarcomaO ProstateO

R

RCCR

Phase II trials

Glioblastoma NSCLC Breast Prostate

Stomach HCCR NSCLCR OvarianR GlioblastomaR Soft tissue carcinomaR ProstateR GlioblastomaR NSCLCR MelanomaR NSCLCR,O MesotheliomaR GISTR PancreasR NeuroendocrineR,O BreastO

RCCR,O

CRCR

CRC: Colorectal carcinoma; GIST: Gastrointestinal stromal tumour; HCC: Hepatocellular carcinoma; O: Ongoing; PDGFR: Platelet-derived growth factor receptor; R: Results available; RCC: Renal cell carcinoma.

HCCO

GISTR,O PancreasO MelanomaO NSCLCR GISTR NSCLCR

CRC: Colorectal carcinoma; GIST: Gastrointestinal stromal tumour; HCC: Hepatocellular carcinoma; O: Ongoing; PDGFR: Platelet-derived growth factor receptor; R: Results available; RCC: Renal cell carcinoma.

mutations. A Phase II trial in non-small cell lung carcinoma and head and neck cancer was discontinued because of blood clot risks. Crenolanib (CP-868,596), in preclinical data, has revealed to be a potent, selective inhibitor of PDGFRb tyrosine kinase, and its 50% inhibitory concentrations is about 1 and 0.4 ng/ ml for a and b receptor types, respectively. It is greater than 100-fold more selective for PDGFR than for other kinases [61]. Crenolanib has been tested in a panel of PDGFRa-mutant kinases expressed in different cell line models, including primary GIST cells, and was significantly more potent than imatinib in inhibiting the kinase activity of imatinib-resistant PDGFRa kinases [34]. Moreover, preclinical data suggest that crenolanib is active against FLT-3 mutation in acute myeloid leukemia [62]. Currently, crenolanib is under development in Phase II trials, especially in GIST with PDGFR deletion. In Phase I trials, dovotinib has been shown to induce adverse events of all grades such as nausea, diarrhea, vomiting, decreased appetite, fatigue, hypertension and hypertriglyceridemia [63]. A Phase II study has also evaluated dovitinib as a

Expert Opin. Investig. Drugs (2015) 24(5)

5

J. Arrondeau et al.

Table 2. IC50 (nM) of PDGFR inhibitors on PDGFRa and PDGFRb.(continued).

Axitinib [33]

Crenolanib [34] Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

Dasatinib [35]

Dovitinib [36]

Imatinib [37]

Lenvatinib [38]

Linifanib [39]

Masitinib [40]

Motesanib [41]

Nilotinib [42]

Nintedanib [43]

Orantinib [44]

Pazopanib [45]

Ponatinib [46]

Regorafenib [47]

Targets

PDGFRa

PDGFRb

Targets

VEGFR PDGFR KIT PDGFR FLT3 BCR-ABL KIT PDGFR SRC EPH A2 FGFR EGFR PDGFR VEGFR PDGFR KIT VEGFR FGFR KIT RET PDGFR PDGFR VEGFR CSF1R FLT3 KIT KIT PDGFR Lyn FGFR VEGFR PDGFR KIT RET BCR-ABL KIT PDGFR VEGFR PDGFR FGFR PDGFR FGFR VEGFR KIT VEGFR PDGFR KIT RAF BCR-ABL RET FGFR KIT PDGFR VEGFR FLT3 TIE2 VEGFR PDGFR

5

1.6

2.1

3.2

KIT RAF FGFR RET VEGFR PDGFR KIT RAF RET FLT3 VEGFR PDGFR KIT RET FLT3 CSF1R VEGFR KIT PDGFR VEGFR PDGFR KIT VEGFR PDGFR

PDGFR: Platelet-derived growth factor receptor.

6

Table 2. IC50 (nM) of PDGFR inhibitors on PDGFRa and PDGFRb.(continued).

Sorafenib [45]

3

210

27

100

-

51

39

Sunitinib [45]

Telatinib [48]

Tivozanib [49] -

66

540

800

Vatalanib [50]

PDGFRa

PDGFRb

933

1129

75

143

-

15

40

49

-

580

PDGFR: Platelet-derived growth factor receptor.

84

71

59

65

-

8

73

215

1.1

7.7

-

22

third-line treatment for GIST and showed modest antitumor activity [64]; others Phase II trials are ongoing with dovitinib alone or in combination with other treatments. Dovitinib has been evaluated in a Phase III trial as a third-line treatment for metastatic renal cell carcinoma and did not show more activity than sorafenib [65]. Lenvatinib has been shown to block migration and invasion of tumor cells with concentrations that inhibit FGFR and PDGFR pathways. Furthermore, lenvatinib could not inhibit cell migration in PDGFR negative cell lines [66]. Phase I and II studies are ongoing with lenvatinib alone or in combination, in different types of tumors without identified PDGFR mutation or overexpression. Recently, a Phase III study has reported an improvement of progression-free survival (PFS) in iodine refractory advanced differentiated thyroid cancer but with major toxicity [67]. It is also currently evaluated in a Phase III trial for first-line treatment of hepatocellular carcinoma versus sorafenib. Linifanib has shown inhibition of PDGFR and c-kit pathways in Ewing sarcoma cells [68]. In Phase II trials, linifanib did not show any improvement of outcome in association with chemotherapy or bevacizumab in colorectal cancer [69], but in lung and hepatocellular carcinoma it has shown activity in monotherapy [70,71]. Others Phase I and II trials are evaluating the efficacy of linifanib, alone or in combination, and a Phase III trial is ongoing in hepatocellular carcinoma comparing its efficacy versus sorafenib.

Expert Opin. Investig. Drugs (2015) 24(5)

Investigational therapies up to Phase II which target PDGF receptors: potential anti-cancer therapeutics

Table 3. Trials with specific PDGFR targeting. Drug

Phase

Status

Indication

Crenolanib Imatinib Nilotinib Olaratumab Ponatinib

II II II II II

Ongoing Suspended Ongoing Ongoing Ongoing

GIST with D842 mutation or PDGFR deletion Glioblastoma expressing PDGFR Melanoma or gliobastoma with PDGFR amplification GIST or glioblastoma with or without PDGFR mutation GIST with KIT or PDGFR mutation

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

GIST: Gastrointestinal stromal tumour; PDGFR: Platelet-derived growth factor receptor.

Masitinib had greater in vitro activity and selectivity against KIT than imatinib and also potently inhibits recombinant PDGFR [40]. In pancreatic cancer, a Phase II trial of masitinib associated with gemcitabine has showed encouraging results [72], leading to an ongoing Phase III trial. Recently, a Phase III study evaluating masitinib as a second-line treatment versus sunitinib in GIST [73] showed a survival benefit; thus, a Phase III trial is ongoing in first-line treatment of advanced or metastatic GIST versus imatinib. Motesanib has demonstrated significant antiangiogenic activity in preclinical data. After a Phase I study [74], a Phase II trial evaluating motesanib in the treatment of ovarian carcinoma was abruptly discontinued because of several cases of posterior reversible encephalopathy syndrome [75]. Recently, patients with squamous cell carcinoma were finally excluded of a Phase III trial evaluating motesanib in lung carcinomas because of limiting toxicities with 47% of serious adverse events, including fatal bleeding events [76]. So far, motesanib has shown modest activity in published data [77-79]. Nintedanib has been shown to consistently reduce the number of pericytes that carry the PDGFR in cell cultures [43] and could provide a possible solution to the observed resistance with other angiogenesis inhibitors [80]. Several Phase I and II trials have also reported its efficacy, alone or in combination, with different results in terms of tumor location, and others are ongoing. Recently, two Phase III studies evaluating nintedanib in combination with chemotherapy have shown OS and PFS improvement in non-small cell lung carcinoma after first-line chemotherapy respectively [81,82]. Orantinib is another tyrosine kinase, which has shown antiangiogenic activity in preclinical data. Despite toxicities reported in the Phase I study [83], orantinib is only under development in a Phase III trial with chemoembolization in patients with unresecable hepatocellular carcinoma. Telatinib has been developed in Phase I studies alone [84,85] or in combination with irinotecan and capecitabine without increased toxicities [86], or with bevacizumab in patients with advanced solid tumors, but this association was very toxic and the development was discontinued [87]. Only one Phase II trial is ongoing in patients with advanced gastric cancer. Tivozanib has shown in Phase I studies that its side effects are generally mild [88] with promising activity in a Phase I study in association with weekly paclitaxel in metastatic breast cancer [89] and in a Phase II trial in combination with

everolimus in colorectal cancer [90]. Recently, a randomized Phase III trial evaluating tivozanib versus sorafenib for firstline treatment of metastatic renal cell carcinoma showed improvement of PFS but not of OS [91]. Vatalanib has been largely developed few years ago, especially in colorectal cancer, with two Phase III randomized trials in combination with chemotherapy, but did not show improvement of outcome. There are also several Phase II trials evaluating its activity alone, in different tumor types but with poor outcomes. Currently, most trials evaluating valatinib are in combination with other treatments in Phase I and II trials, with chemotherapy, or with other targeted therapy.

Phase I and II trials of monoclonal antibodies which target PDGFR

3.2

Monoclonal antibodies have more specific targeting than tyrosine kinase inhibitors and have therefore usually less toxicity. In addition, they could lead to an immune response including complement-dependent cytotoxicity and antibody-dependent cellular cytotoxicity. Two antibodies targeting selectively PDGFRa, and no cross-reacting with PDGFRb, have been developed. In preclinical data, antibodies showed efficacy in tumor models dependent on PDGFRa signaling [92,93]. Olaratumab (IMC-3G3) is a fully human IgG1 mAb. In the Phase I trials, the most common adverse events were fatigue and infusion reaction, and olaratumab showed the preliminary antitumor activity [94]. Two Phase II trials evaluating olaratumab as monotherapy have been completed in GIST with or without PDGFRa mutations and in glioblastomas. In mouse models of ovarian tumors, high expression of PDGFRa was correlated with poor survival and olaratumab was only effective in combination with docetaxel, and thus could lead to sensitization to chemotherapy [95]. Therefore, olaratumab is specially developed in combination with other treatments and several trials are currently ongoing: a Phase II study evaluating olaratumab in association with carboplatin and paclitaxel in non-small cell lung carcinoma, a Phase II trial in association with liposomal doxorubicin in platinium-refractory ovarian cancer, a Phase I/II study in association with doxorubicin in soft tissue sarcomas, and a Phase II study in association with mitoxantrone in prostate cancer. Tovetumab (MEDI575) is a human IgG2 k mAb directed against PDGFRa. A Phase I trial reported fatigue and nausea

Expert Opin. Investig. Drugs (2015) 24(5)

7

X

X

X

X

X X

X

Dasatinib

PDGFR: Platelet-derived growth factor receptor.

Fatigue Anorexia Pain Pyrexia Nausea/vomiting Diarrhea Cytolysis Pancreatis Stomatitis Dyspnea Hematologic Hypertension PRESS syndrome Thromboembolic Hemorrhage Palmoplantar syndrome Rash Pleural/Pericardic effusion Hypertryglyceridemia

Crenolanib

X

X

X X

X X

Dovitinib

X

X

X X

Lenvatinib

X

X

X X

X X

Linifanib

X

X

X X

X X

X X X

X

X

Motesanib

X

Masitinib

Table 4. Main adverse events reported with PDGFR targeted molecules under development.

X

X

X

X

Nilotinib

X X X

Nintedanib

X

X X X

Orantinib

X

X

X

X

X

X

Ponatinib

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

X

X X

Telatinib

X

X

X

Tivozanib

X

X

X

X

Vatalanib

Investigational therapies up to Phase II which target PDGF receptors: potential anti-cancer therapeutics

as most frequent adverse events, but did not show evidence of tumor activity [96]. Two Phase II trials have been completed, in glioblastoma and in non-small cell lung carcinoma in combination with chemotherapy, but development has recently been discontinued.

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

4.

Conclusion

The PDFGR pathway has important functions in normal cell growth and, by overexpression or mutations, can lead to tumor development. Moreover PDGFR is expressed in the tumor microenvironment and could promote tumorigenesis. This review reports preclinical and clinical rationale for targeting PDGFR as anti-cancer therapy. Many agents targeting PDGFR are currently under development, most of them being non-selective tyrosine kinase inhibitors, whereas two are monoclonal antibodies. The status of development of theses different molecules is varied and in heterogeneous types of tumors. Nevertheless those therapies are not expected to have a major global anti-tumor effect and many could fail to become approved therapies. Improvement in the development of PDGFR inhibitors is needed. 5.

Expert opinion

This comprehensive report reviews the role of PDGFR pathway in tumorigenesis and the clinical development of different PDGFR inhibitors. PDGFR has been a target for the development of therapy in medicine since the 1990s and a bit of history may be interesting. At that time the objective was to develop molecules having the ability to inhibit PDGFR, EGFR and Protein Kinase C, which could be of use in treating fibrosis, atherosclerosis and restenosis. Different molecules emerged from this kinase inhibitors program, which was mostly dedicated to the treatment of cardiovascular disease. At the same time, BCR-ABL protein dysregulation was identified as the event leading to white blood cells proliferation in CML and existing compounds were screened for their ability to inhibit BCR-ABL [97]. This is how imatinib, initially developed as a PDGFR inhibitor, became the first targeted therapy in oncology. Furthermore in 2001 although imatinib had started its development in CML, a first case report evaluating imatinib in a patient with a GIST and showing a complete response was published [98]. After this publication, development of imatinib in GIST was initiated and only after PDGFR mutations in GIST were discovered. This historical example highlights that imatinib, the first targeted therapy, which was originally developed to inhibit PDGFR, has mainly been used in settings where the inhibition of PDGFR was not the target. In our opinion, the key messages of this review are the following. First, currently approved PDGFR inhibitors exist, but their targeting is not specific of PDGFR as they are multityrosine kinase inhibitors; their safety profile is now well determined and their toxicity is mostly manageable like for

imatinib. Second, despite preclinical data showing an important role of PDGFR in tumorigenesis, those PDGFR inhibitors are mostly prescribed not for their ability to inhibit PDGFR but because they target others tyrosine kinases mostly bcr-abl, kit, VEGFR; in fact the targeting of PDGFR in solid tumors is currently limited to GIST and DFSP. Third, only a limited number of molecules inhibiting PDGFR currently under development are indeed developed for their ability to inhibit PDGFR. So how those investigational therapies could be used as anti-cancer therapies remains unclear. A few rules that apply across all anti-cancer therapies should be reminded. 1) Having determined the target of the treatment is nowadays crucial for the development of anti-cancer therapeutics as predictive biomarkers of outcome are explored in large Phase III trials. This aspect is also a growing demand of regulators when approving an anti-cancer therapy. The key of success for a targeted therapy is the identification of an oncogenic driver as target. In lung cancer for example, EGFR inhibitors have significant efficacy in EGFR mutated lung cancers [99-101], because EGFR mutations are drivers of the disease [102]. In GIST, DFSP, and some leukemias, PDGFR is an important driver of the disease [103-105] and targeting PDGFR is effective. For these reasons, molecules and studies highlighted in Table 3 have a real chance to become anti-cancer therapies and this is particularly true when they are developed in tumors with unmet therapeutic needs such as glioblastomas. A new method, the study of the kinome (the activation of different tyrosine kinases) could help determine if the molecule under investigation is actually targeting the right target across all patients. In our institution a study is currently ongoing for patients treated with sunitinib for whom lymphocytes tyrosine kinases activity is determined prior and under treatment to determine the in vivo effect of the therapy. 2) A high selectivity in the targeting of an investigational agent can be wished or not but this has to be determined first. A highly selective agent is expected to have limited off-target adverse effects and a good safety profile and could therefore be developed either in a cancer with a specific activation of the pathway resulting from a limited number of mutations or in combination therapy. This point is crucial as two recent concepts are in favor of the inhibition of multiple pathways. The concept of synthetic lethality [106] implying the combination of two or more therapies leading to cancer-cell death whereas a single therapy would not, requires to have limited toxicity and non-overlapping toxicities. The concept of biological crosstalk refers to instances in which one or more components of one signal transduction pathway affects another signal

Expert Opin. Investig. Drugs (2015) 24(5)

9

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

J. Arrondeau et al.

transduction pathway. This can be achieved with shared components that can interact with either pathway. Those two concepts are important for the development of PDGFR inhibitors as for most tumor types the PDGFR pathway is not the only driver of tumor growth. Therefore the inhibition of PDGFR has to be associated to the inhibition of another pathway. This can be achieved with a multi-targeted tyrosine kinase inhibitor or with a very specific inhibitor associated to a specific inhibitor of another pathway. New engineered monoclonal antibodies appear to be useful in this setting. The drawback of a high selectivity could be an early mechanism of resistance from the cancer cells and therefore a limited efficacy when a wide targeting of tyrosine kinases and cellular pathways is more difficult to bypass. But of course clinically limiting toxicities are to be expected. 3) A sustained optimal exposition to the investigational therapy must be guaranteed to determine its efficacy. Recent publications have shown significant intra- and inter-individual variations in tyrosine kinase inhibitors bioavailability. Indeed, it has been demonstrated that exposure to sorafenib decreases over time [107] and that in some settings, dose escalation can restore efficacy [108,109]. For this reason the development of novel tyrosine kinase inhibitor must include an evaluation of the plasmatic exposure to the agent. This can be done by searching on-target adverse events for patients as it has been done for axitinib in a trial versus sorafenib in metastatic renal cell cancer [110]. But a more objective and patient-centered method is to simply determine the blood concentration. As a result, a trial comparing an approved therapy given at a flat dose to a novel molecule having the same molecular targets and safety profile but with blood drug monitoring to guarantee a sustained active exposure is expected to show significant benefit in antitumor activity with the challenger. But it has to be added that some particularities of PDGFR inhibition must be taken into account for the development PDGFR inhibitors. Because tumor cells and stroma cells associated to solid tumors express PDGFR, targeting PDGFR will affect both normal and cancer cells. In the PDGFR-negative tumor cell, the impact of an anti-PDGF drug that exclusively targets the stromal compartment can be studied. A preclinical study hypothesized that the overexpression of PDGF-BB in PDGFR-negative colorectal cancer and pancreatic cancer cells would result in increased pericyte coverage of endothelial cells in vivo, resulting in the tumor vasculature more resistant to antiangiogenic therapy. Treatment of PDGF-BB-overexpressing tumors with imatinib resulted in increased growth and

10

decreased total pericyte content compared with those in untreated PDGF-BB-overexpressing tumors suggesting that PDGFR inhibitor agents should be used with caution when PDGFR is not the target on the cancer-cell itself [111]. Moreover the PDGFR pathway is closely entangled with other pathways, such as VEGFR, EGFR, FGFR, MET, and KIT. Thus, multikinase inhibitors could have interest in the approach of targeting PDGFR. An example in GISTs is that PDGFRa and KIT seem to be mutually exclusive and one mutated receptor can heterodimerize with the wild-type version of the other receptor, producing diverse signaling [103]. In this case, drugs such imatinib or sunitinib targeting both Kit and PDGFRa pathways would be more effective than monospecific targeted ones. Moreover, combination of anti-PDGFR and antiVEGFR inhibitors could have activity, because anti-PDGFR drug would induce ablation of pericytes leaving the tumor vessels accessible for anti-VEGFR drug. In preclinical data simultaneous inhibition of VEGFR and PDGFR with the kinase inhibitor SU6668 markedly sensitized xenograft tumor response to radiation therapy [112]. Interestingly, resistance to anti-VEGF treatment has been shown to involve PDGF-AA, -BB and --CC [113]. More agents targeting PDGFR are currently under development. Some seem to have the potential to become anticancer therapies given their targeting or their safety profile; however their development may prevent this from happening. In our opinion, whenever possible, studies should be based on a PDGFR over-activated pathway in cancer cells as preclinical studies indicate that PDGFR inhibition in PDGFR-negative tumor cells would result in tumor growth. Agents with less inter- and intra- individual variability in bioavailability should be preferred. Drug monitoring should be associated to the development to guarantee a sustained active exposure. A highly selective agent with limited toxicity, such as an mAb, could be important for combination therapies. If not highly selective, the agent should also target VEGFR as the effect is supposed to be synergistic with the inhibition of PDGFR. Finally current developers in highly specialized fields such as neuro-oncology should integrate in their development plan what has been already demonstrated across the oncology field during the last decade.

Declaration of interest The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Expert Opin. Investig. Drugs (2015) 24(5)

Investigational therapies up to Phase II which target PDGF receptors: potential anti-cancer therapeutics

Bibliography

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

Papers of special note have been highlighted as either of interest () or of considerable interest () to readers. 1.

Fredriksson L, Li H, Eriksson U. The PDGF family: four gene products form five dimeric isoforms. Cytokine Growth Factor Rev 2004;15(4):197--204

2.

Uutela M, Lauren J, Bergsten E, et al. Chromosomal location, exon structure, and vascular expression patterns of the human PDGFC and PDGFD genes. Circulation 2001;103(18):2242--7

3.

.

4.

5.

6.

Andrae J, Gallini R, Betsholtz C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev 2008;22(10):1276--312 This article explains with precision the role of platelet-derived growth factor receptor (PDGFR) in cellular biology. Roberts WG, Whalen PM, Soderstrom E, et al. Antiangiogenic and antitumor activity of a selective PDGFR tyrosine kinase inhibitor, CP-673,451. Cancer Res 2005;65(3):957--66 Oikawa T, Onozawa C, Sakaguchi M, et al. Three isoforms of platelet-derived growth factors all have the capability to induce angiogenesis in vivo. Biol Pharm Bull 1994;17(12):1686--8 Betsholtz C. Insight into the physiological functions of PDGF through genetic studies in mice. Cytokine Growth Factor Rev 2004;15(4):215--28

7.

Jechlinger M, Sommer A, Moriggl R, et al. Autocrine PDGFR signaling promotes mammary cancer metastasis. J Clin Invest 2006;116(6):1561--70

8.

Heinrich MC, Corless CL, Duensing A, et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science 2003;299(5607):708--10 This article demonstrates that PDGFR could be an oncogenic driver in gastrointestinal stromal tumor (GIST), which has therapeutic consequences.

.

9.

10.

Puputti M, Tynninen O, Sihto H, et al. Amplification of KIT, PDGFRA, VEGFR2, and EGFR in gliomas. Mol Cancer Res 2006;4(12):927--34 Shimizu A, O’Brien KP, Sj€oblom T, et al. The dermatofibrosarcoma protuberans-associated collagen type Ialpha1/platelet-derived growth factor (PDGF) B-chain fusion gene generates a transforming protein that is processed to

carcinoma. Clin Cancer Res 2006;12(9):2676--88

functional PDGF-BB. Cancer Res 1999;59(15):3719--23 11.

Jones AV, Cross NCP. Oncogenic derivatives of platelet-derived growth factor receptors. Cell Mol Life Sci 2004;61(23):2912--23

12.

Ustach CV, Taube ME, Hurst NJ, et al. A potential oncogenic activity of plateletderived growth factor d in prostate cancer progression. Cancer Res 2004;64(5):1722--9

13.

Fischer ANM, Fuchs E, Mikula M, et al. PDGF essentially links TGF-beta signaling to nuclear beta-catenin accumulation in hepatocellular carcinoma progression. Oncogene 2007;26(23):3395--405

14.

Kawai T, Hiroi S, Torikata C. Expression in lung carcinomas of platelet-derived growth factor and its receptors. Lab Invest 1997;77(5):431--6

15.

Bhardwaj B, Klassen J, Cossette N, et al. Localization of platelet-derived growth factor beta receptor expression in the periepithelial stroma of human breast carcinoma. Clin Cancer Res 1996;2(4):773--82

16.

Henriksen R, Funa K, Wilander E, et al. Expression and prognostic significance of platelet-derived growth factor and its receptors in epithelial ovarian neoplasms. Cancer Res 1993;53(19):4550--4

17.

Sundberg C, Ljungstr€om M, Lindmark G, et al. Microvascular pericytes express platelet-derived growth factor-beta receptors in human healing wounds and colorectal adenocarcinoma. Am J Pathol 1993;143(5):1377--88

18.

Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 2012;21(3):309--22 This review shows that tumor microenvironment could have a real importance in tumor growth; thus therapeutic development should include tumor microenvironment.

..

19.

Shao ZM, Nguyen M, Barsky SH. Human breast carcinoma desmoplasia is PDGF initiated. Oncogene 2000;19(38):4337--45

20.

Tejada ML, Yu L, Dong J, et al. Tumor-driven paracrine platelet-derived growth factor receptor alpha signaling is a key determinant of stromal cell recruitment in a model of human lung

Expert Opin. Investig. Drugs (2015) 24(5)

21.

Campbell JS, Hughes SD, Gilbertson DG, et al. Platelet-derived growth factor C induces liver fibrosis, steatosis, and hepatocellular carcinoma. Proc Natl Acad Sci USA 2005;102(9):3389--94

22.

Anderberg C, Li H, Fredriksson L, et al. Paracrine signaling by platelet-derived growth factor-CC promotes tumor growth by recruitment of cancerassociated fibroblasts. Cancer Res 2009;69(1):369--78

23.

Bandapalli OR, Macher-Goeppinger S, Schirmacher P, Brand K. Paracrine signalling in colorectal liver metastases involving tumor cell-derived PDGF-C and hepatic stellate cell-derived PAK-2. Clin Exp Metastasis 2012;29(5):409--17

24.

Strawn LM, Mann E, Elliger SS, et al. Inhibition of glioma cell growth by a truncated platelet-derived growth factorbeta receptor. J Biol Chem 1994;269(33):21215--22

25.

Uehara H, Kim SJ, Karashima T, et al. Effects of blocking platelet-derived growth factor-receptor signaling in a mouse model of experimental prostate cancer bone metastases. J Natl Cancer Inst 2003;95(6):458--70

26.

Garofalo A, Naumova E, Manenti L, et al. The combination of the tyrosine kinase receptor inhibitor SU6668 with paclitaxel affects ascites formation and tumor spread in ovarian carcinoma xenografts growing orthotopically. Clin Cancer Res 2003;9(9):3476--85

27.

Machida S, Saga Y, Takei Y, et al. Inhibition of peritoneal dissemination of ovarian cancer by tyrosine kinase receptor inhibitor SU6668 (TSU-68). Int J Cancer 2005;114(2):224--9

28.

Sj€oblom T, Shimizu A, O’Brien KP, et al. Growth inhibition of dermatofibrosarcoma protuberans tumors by the platelet-derived growth factor receptor antagonist STI571 through induction of apoptosis. Cancer Res 2001;61(15):5778--83

29.

Xi Y, Chen M, Liu X, et al. CP-673451, a platelet-derived growth-factor receptor inhibitor, suppresses lung cancer cell proliferation and migration. Onco Targets Ther 2014;7:1215--21

11

J. Arrondeau et al.

30.

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

.

31.

32.

33.

34.

35.

36.

37.

38.

12

Corless CL, Schroeder A, Griffith D, et al. PDGFRA mutations in gastrointestinal stromal tumors: frequency, spectrum and in vitro sensitivity to imatinib. J Clin Oncol 2005;23(23):5357--64 This article is one the first showing efficacy of imatinib as a targeted therapy for PDGFR mutations in GIST. Mathew P, Thall PF, Wen S, et al. Dynamic change in phosphorylated platelet-derived growth factor receptor in peripheral blood leukocytes following docetaxel therapy predicts progressionfree and overall survival in prostate cancer. Br J Cancer 2008;99(9):1426--32 Matei D, Emerson RE, Schilder J, et al. Imatinib mesylate in combination with docetaxel for the treatment of patients with advanced, platinum-resistant ovarian cancer and primary peritoneal carcinomatosis : a Hoosier Oncology Group trial. Cancer 2008;113(4):723--32 Hu-Lowe DD, Zou HY, Grazzini ML, et al. Nonclinical antiangiogenesis and antitumor activities of axitinib (AG013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptor tyrosine kinases 1, 2, 3. Clin Cancer Res 2008;14(22):7272--83 Heinrich MC, Griffith D, McKinley A, et al. Crenolanib inhibits the drugresistant PDGFRA D842V mutation associated with imatinib-resistant gastrointestinal stromal tumors. Clin Cancer Res 2012;18(16):4375--84 Chen Z, Lee FY, Bhalla KN, Wu J. Potent inhibition of platelet-derived growth factor-induced responses in vascular smooth muscle cells by BMS354825 (dasatinib). Mol Pharmacol 2006;69(5):1527--33

cancer H146, based on angiogenesis inhibition. Int J Cancer 2008;122(3):664--71 39.

Albert DH, Tapang P, Magoc TJ, et al. Preclinical activity of ABT-869, a multitargeted receptor tyrosine kinase inhibitor. Mol Cancer Ther 2006;5(4):995--1006

40.

Dubreuil P, Letard S, Ciufolini M, et al. Masitinib (AB1010), a potent and selective tyrosine kinase inhibitor targeting KIT. PLoS One 2009;4(9):e7258

41.

42.

43.

44.

Matsui J, Yamamoto Y, Funahashi Y, et al. E7080, a novel inhibitor that targets multiple kinases, has potent antitumor activities against stem cell factor producing human small cell lung

Weisberg E, Manley PW, Breitenstein W, et al. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell 2005;7(2):129--41 Hilberg F, Roth GJ, Krssak M, et al. BIBF 1120: triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Res 2008;68(12):4774--82 Laird AD, Vajkoczy P, Shawver LK, et al. SU6668 is a potent antiangiogenic and antitumor agent that induces regression of established tumors. Cancer Res 2000;60(15):4152--60

49.

Nakamura K, Taguchi E, Miura T, et al. KRN951, a highly potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, has antitumor activities and affects functional vascular properties. Cancer Res 2006;66(18):9134--42

50.

Wood JM, Bold G, Buchdunger E, et al. PTK787/ZK 222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, impairs vascular endothelial growth factorinduced responses and tumor growth after oral administration. Cancer Res 2000;60(8):2178--89

51.

Mayer EL, Baurain J-F, Sparano J, et al. A phase 2 trial of dasatinib in patients with advanced HER2-positive and/or hormone receptor-positive breast cancer. Clin Cancer Res 2011;17(21):6897--904

52.

Finn RS, Bengala C, Ibrahim N, et al. Dasatinib as a single agent in triplenegative breast cancer: results of an openlabel phase 2 study. Clin Cancer Res 2011;17(21):6905--13

53.

Schilder RJ, Brady WE, Lankes HA, et al. Phase II evaluation of dasatinib in the treatment of recurrent or persistent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol 2012;127(1):70--4

54.

Twardowski PW, Beumer JH, Chen CS, et al. A phase II trial of dasatinib in patients with metastatic castrationresistant prostate cancer treated previously with chemotherapy. Anticancer Drugs 2013;24(7):743--53

45.

Kumar R, Crouthamel M-C, Rominger DH, et al. Myelosuppression and kinase selectivity of multikinase angiogenesis inhibitors. Br J Cancer 2009;101(10):1717--23

55.

46.

O’Hare T, Shakespeare WC, Zhu X, et al. AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell 2009;16(5):401--12

Araujo JC, Trudel GC, Paliwal P. Long-term use of dasatinib in patients with metastatic castration-resistant prostate cancer after receiving the combination of dasatinib and docetaxel. Cancer Manag Res 2013;6:25--30

56.

Brunner AM, Costa DB, Heist RS, et al. Treatment-related toxicities in a phase II trial of dasatinib in patients with squamous cell carcinoma of the lung. J Thorac Oncol 2013;8(11):1434--7

57.

Chee CE, Krishnamurthi S, Nock CJ, et al. Phase II study of dasatinib (BMS354825) in patients with metastatic adenocarcinoma of the pancreas. Oncologist 2013;18(10):1091--2

Trudel S, Li ZH, Wei E, et al. CHIR258, a novel, multitargeted tyrosine kinase inhibitor for the potential treatment of t(4;14) multiple myeloma. Blood 2005;105(7):2941--8 Heinrich MC, Corless CL, Demetri GD, et al. Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J Clin Oncol 2003;21(23):4342--9

Polverino A, Coxon A, Starnes C, et al. AMG 706, an oral, multikinase inhibitor that selectively targets vascular endothelial growth factor, platelet-derived growth factor, and kit receptors, potently inhibits angiogenesis and induces regression in tumor xenografts. Cancer Res 2006;66(17):8715--21

molecule angiogenesis inhibitor. Clin Cancer Res 2008;14(11):3470--6

47.

48.

Wilhelm SM, Dumas J, Adnane L, et al. Regorafenib (BAY 73-4506): a new oral multikinase inhibitor of angiogenic, stromal and oncogenic receptor tyrosine kinases with potent preclinical antitumor activity. Int J Cancer 2011;129(1):245--55 Steeghs N, Gelderblom H, Roodt JO, et al. Hypertension and rarefaction during treatment with telatinib, a small

Expert Opin. Investig. Drugs (2015) 24(5)

Investigational therapies up to Phase II which target PDGF receptors: potential anti-cancer therapeutics

58.

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

59.

60.

61.

62.

63.

64.

65.

Araujo JC, Trudel GC, Saad F, et al. Docetaxel and dasatinib or placebo in men with metastatic castration-resistant prostate cancer (READY): a randomised, double-blind phase 3 trial. Lancet Oncol 2013;14(13):1307--16 Weigel MT, Rath K, Alkatout I, et al. Nilotinib in combination with carboplatin and paclitaxel is a candidate for ovarian cancer treatment. Oncology 2014;87(4):232--45 Kanda T, Ishikawa T, Takahashi T, Nishida T. Nilotinib for treatment of gastrointestinal stromal tumors: out of the equation? Expert Opin Pharmacother 2013;14(13):1859--67 Lewis NL, Lewis LD, Eder JP, et al. Phase I study of the safety, tolerability, and pharmacokinetics of oral CP868,596, a highly specific platelet-derived growth factor receptor tyrosine kinase inhibitor in patients with advanced cancers. J Clin Oncol 2009;27(31):5262--9 Zimmerman EI, Turner DC, Buaboonnam J, et al. Crenolanib is active against models of drug-resistant FLT3-ITD-positive acute myeloid leukemia. Blood 2013;122(22):3607--15 Angevin E, Lopez-Martin JA, Lin C-C, et al. Phase I study of dovitinib (TKI258), an oral FGFR, VEGFR, and PDGFR inhibitor, in advanced or metastatic renal cell carcinoma. Clin Cancer Res 2013;19(5):1257--68 Kang Y-K, Yoo C, Ryoo B-Y, et al. Phase II study of dovitinib in patients with metastatic and/or unresectable gastrointestinal stromal tumours after failure of imatinib and sunitinib. Br J Cancer 2013;109(9):2309--15 Motzer RJ, Porta C, Vogelzang NJ, et al. Dovitinib versus sorafenib for third-line targeted treatment of patients with metastatic renal cell carcinoma: an openlabel, randomised phase 3 trial. Lancet Oncol 2014;15(3):286--96

66.

Glen H, Mason S, Patel H, et al. E7080, a multi-targeted tyrosine kinase inhibitor suppresses tumor cell migration and invasion. BMC Cancer 2011;11:309

67.

Lenvatinib slows progression of thyroid cancer. Cancer Discov 2014;4(8):OF7

68.

Ikeda AK, Judelson DR, Federman N, et al. ABT-869 inhibits the proliferation of Ewing Sarcoma cells and suppresses platelet-derived growth factor receptor

beta and c-KIT signaling pathways. Mol Cancer Ther 2010;9(3):653--60

78.

Benjamin RS, Sch€offski P, Hartmann JT, et al. Efficacy and safety of motesanib, an oral inhibitor of VEGF, PDGF, and Kit receptors, in patients with imatinibresistant gastrointestinal stromal tumors. Cancer Chemother Pharmacol 2011;68(1):69--77

79.

Toh HC, Chen P-J, Carr BI, et al. Phase 2 trial of linifanib (ABT-869) in patients with unresectable or metastatic hepatocellular carcinoma. Cancer 2013;119(2):380--7

Schlumberger MJ, Elisei R, Bastholt L, et al. Phase II study of safety and efficacy of motesanib in patients with progressive or symptomatic, advanced or metastatic medullary thyroid cancer. J Clin Oncol 2009;27(23):3794--801

80.

71.

Tan E-H, Goss GD, Salgia R, et al. Phase 2 trial of Linifanib (ABT-869) in patients with advanced non-small cell lung cancer. J Thorac Oncol 2011;6(8):1418--25

Casanovas O, Hicklin DJ, Bergers G, Hanahan D. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 2005;8(4):299--309

81.

72.

Mitry E, Hammel P, Deplanque G, et al. Safety and activity of masitinib in combination with gemcitabine in patients with advanced pancreatic cancer. Cancer Chemother Pharmacol 2010;66(2):395--403

Reck M, Kaiser R, Mellemgaard A, et al. Docetaxel plus nintedanib versus docetaxel plus placebo in patients with previously treated non-small-cell lung cancer (LUME-Lung 1): a phase 3, double-blind, randomised controlled trial. Lancet Oncol 2014;15(2):143--55

73.

Adenis A, Blay J-Y, Bui-Nguyen B, et al. Masitinib in advanced gastrointestinal stromal tumor (GIST) after failure of imatinib: a randomized controlled openlabel trial. Ann Oncol 2014;25(9):1762--9

82.

74.

Rosen LS, Kurzrock R, Mulay M, et al. Safety, pharmacokinetics, and efficacy of AMG 706, an oral multikinase inhibitor, in patients with advanced solid tumors. J Clin Oncol 2007;25(17):2369--76

Hanna NH, Kaiser R, Sullivan RN, et al. Lume-lung 2: a multicenter, randomized, double-blind, phase III study of nintedanib plus pemetrexed versus placebo plus pemetrexed in patients with advanced nonsquamous non-small cell lung cancer (NSCLC) after failure of first-line chemotherapy. J Clin Oncol 2013;31(Suppl):abstract 8034

83.

Kuenen BC, Giaccone G, Ruijter R, et al. Dose-finding study of the multitargeted tyrosine kinase inhibitor SU6668 in patients with advanced malignancies. Clin Cancer Res 2005;11(17):6240--6

84.

Strumberg D, Schultheis B, Adamietz IA, et al. Phase I dose escalation study of telatinib (BAY 57-9352) in patients with advanced solid tumours. Br J Cancer 2008;99(10):1579--85

85.

Eskens FALM, Steeghs N, Verweij J, et al. Phase I dose escalation study of telatinib, a tyrosine kinase inhibitor of vascular endothelial growth factor receptor 2 and 3, platelet-derived growth factor receptor beta, and c-Kit, in patients with advanced or metastatic solid tumors. J Clin Oncol 2009;27(25):4169--76

86.

Langenberg MHG, Witteveen PO, Roodhart J, et al. Phase I evaluation of telatinib, a VEGF receptor tyrosine kinase inhibitor, in combination with

69.

70.

75.

76.

77.

O’Neil BH, Cainap C, Van Cutsem E, et al. Randomized phase II open-label study of mFOLFOX6 in combination with linifanib or bevacizumab for metastatic colorectal cancer. Clin Colorectal Cancer 2014;13(3):156--163.e2

Schilder RJ, Sill MW, Lankes HA, et al. A phase II evaluation of motesanib (AMG 706) in the treatment of persistent or recurrent ovarian, fallopian tube and primary peritoneal carcinomas: a Gynecologic Oncology Group study. Gynecol Oncol 2013;129(1):86--91 Novello S, Scagliotti GV, Sydorenko O, et al. Motesanib plus carboplatin/ paclitaxel in patients with advanced squamous non-small-cell lung cancer: results from the randomized controlled MONET1 study. J Thorac Oncol 2014;9(8):1154--61 Scagliotti GV, Vynnychenko I, Park K, et al. International, randomized, placebocontrolled, double-blind phase III study of motesanib plus carboplatin/paclitaxel in patients with advanced nonsquamous non-small-cell lung cancer: MONET1. J Clin Oncol 2012;30(23):2829--36

Expert Opin. Investig. Drugs (2015) 24(5)

13

J. Arrondeau et al.

monoclonal antibody, in patients with advanced solid tumors. Cancer Chemother Pharmacol 2014;73(3):595--604

bevacizumab in subjects with advanced solid tumors. Ann Oncol 2011;22(11):2508--15

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

87.

88.

Langenberg MHG, Witteveen PO, Roodhart JM, et al. Phase I evaluation of telatinib, a vascular endothelial growth factor receptor tyrosine kinase inhibitor, in combination with irinotecan and capecitabine in patients with advanced solid tumors. Clin Cancer Res 2010;16(7):2187--97 Eskens FALM, de Jonge MJA, Bhargava P, et al. Biologic and clinical activity of tivozanib (AV-951, KRN951), a selective inhibitor of VEGF receptor-1, -2, and -3 tyrosine kinases, in a 4-week-on, 2-week-off schedule in patients with advanced solid tumors. Clin Cancer Res 2011;17(22):7156--63

95.

96.

Matsuo K, Nishimura M, Komurov K, et al. Platelet-derived growth factor receptor alpha (PDGFRa) targeting and relevant biomarkers in ovarian carcinoma. Gynecol Oncol 2014;132(1):166--75 Becerra CR, Conkling P, Vogelzang N, et al. A phase I dose-escalation study of MEDI-575, a PDGFRa monoclonal antibody, in adults with advanced solid tumors. Cancer Chemother Pharmacol 2014;74(5):917--25

97.

Druker BJ. Translation of the Philadelphia chromosome into therapy for CML. Blood 2008;112(13):4808--17

89.

Mayer EL, Scheulen ME, Beckman J, et al. A Phase I dose-escalation study of the VEGFR inhibitor tivozanib hydrochloride with weekly paclitaxel in metastatic breast cancer. Breast Cancer Res Treat 2013;140(2):331--9

98.

Joensuu H, Roberts PJ, Sarlomo-Rikala M, et al. Effect of the tyrosine kinase inhibitor STI571 in a patient with a metastatic gastrointestinal stromal tumor. N Engl J Med 2001;344(14):1052--6

90.

Wolpin BM, Ng K, Zhu AX, et al. Multicenter phase II study of tivozanib (AV-951) and everolimus (RAD001) for patients with refractory, metastatic colorectal cancer. Oncologist 2013;18(4):377--8

99.

Rosell R, Moran T, Queralt C, et al. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med 2009;361(10):958--67

91.

Motzer RJ, Nosov D, Eisen T, et al. Tivozanib versus sorafenib as initial targeted therapy for patients with metastatic renal cell carcinoma: results from a phase III trial. J Clin Oncol 2013;31(30):3791--9

92.

Gerber DE, Gupta P, Dellinger MT, et al. Stromal platelet-derived growth factor receptor a (PDGFRa) provides a therapeutic target independent of tumor cell PDGFRa expression in lung cancer xenografts. Mol Cancer Ther 2012;11(11):2473--82 An article that underlines the importance of tumor microenvironment.

..

93.

94.

14

Laing N, McDermott B, Wen S, et al. Inhibition of platelet-derived growth factor receptor a by MEDI-575 reduces tumor growth and stromal fibroblast content in a model of non-small cell lung cancer. Mol Pharmacol 2013;83(6):1247--56 Chiorean EG, Sweeney C, Youssoufian H, et al. A phase I study of olaratumab, an anti-platelet-derived growth factor receptor alpha (PDGFRa)

100. Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutationpositive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 2012;13(3):239--46 .. A cornerstone article in the development of EGFR tyrosine kinase inhibitor for NSCLC treatment. 101. Wu Y-L, Zhou C, Hu C-P, et al. Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): an open-label, randomised phase 3 trial. Lancet Oncol 2014;15(2):213--22 102. Sordella R, Bell DW, Haber DA, Settleman J. Gefitinib-sensitizing EGFR mutations in lung cancer activate antiapoptotic pathways. Science 2004;305(5687):1163--7 103. Negri T, Bozzi F, Conca E, et al. Oncogenic and ligand-dependent activation of KIT/PDGFRA in surgical samples of imatinib-treated

Expert Opin. Investig. Drugs (2015) 24(5)

gastrointestinal stromal tumours (GISTs). J Pathol 2009;217(1):103--12 104. Sawyers CL. Imatinib GIST keeps finding new indications: successful treatment of dermatofibrosarcoma protuberans by targeted inhibition of the platelet-derived growth factor receptor. J Clin Oncol 2002;20(17):3568--9 105. Golub TR, Barker GF, Lovett M, Gilliland DG. Fusion of PDGF receptor beta to a novel ets-like gene, tel, in chronic myelomonocytic leukemia with t (5;12) chromosomal translocation. Cell 1994;77(2):307--16 106. McLornan DP, List A, Mufti GJ. Applying synthetic lethality for the selective targeting of cancer. N Engl J Med 2014;371(18):1725--35 107. Arrondeau J, Mir O, Boudou-Rouquette P, et al. Sorafenib exposure decreases over time in patients with hepatocellular carcinoma. Invest New Drugs 2012;30(5):2046--9 .. These three last references [107-109] highlight that drug exposure monitoring could have a critical role in safety and antitumor activity. 108. Bellesoeur A, Carton E, Mir O, et al. Critical role of sorafenib exposure over time for its antitumor activity in thyroid cancer. Invest New Drugs 2014;32(3):569--72 .. These three last references [107-109] highlight that drug exposure monitoring could have a critical role in safety and antitumor activity. 109. Huillard O, Boissier E, Blanchet B, et al. Drug safety evaluation of sorafenib for treatment of solid tumors: consequences for the risk assessment and management of cancer patients. Expert Opin Drug Saf 2014;13(5):663--73 .. These three last references [107-109] highlight that drug exposure monitoring could have a critical role in safety and antitumor activity. 110. Hutson TE, Lesovoy V, Al-Shukri S, et al. Axitinib versus sorafenib as firstline therapy in patients with metastatic renal-cell carcinoma: a randomised openlabel phase 3 trial. Lancet Oncol 2013;14(13):1287--94 111. McCarty MF, Somcio RJ, Stoeltzing O, et al. Overexpression of PDGF-BB decreases colorectal and pancreatic cancer growth by increasing tumor pericyte content. J Clin Invest 2007;117(8):2114--22

Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by Biblioteka Uniwersytetu Warszawskiego on 02/13/15 For personal use only.

Investigational therapies up to Phase II which target PDGF receptors: potential anti-cancer therapeutics

112.

Griffin RJ, Williams BW, Wild R, et al. Simultaneous inhibition of the receptor kinase activity of vascular endothelial, fibroblast, and platelet-derived growth factors suppresses tumor growth and enhances tumor radiation response. Cancer Res 2002;62(6):1702--6

113.

Crawford Y, Kasman I, Yu L, et al. PDGF-C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to antiVEGF treatment. Cancer Cell 2009;15(1):21--34

Affiliation Jennifer Arrondeau1, Olivier Huillard1, Camille Tlemsani1, Anatole Cessot1, Pascaline Boudou-Rouquette1, Benoit Blanchet2, Audrey Thomas-Schoemann2, Michel Vidal2, Jean-Marie Tigaud1, Jean-Philippe Durand1, Jerome Alexandre1 & Francois Goldwasser†1 † Author for correspondence 1 Paris Descartes University, Cochin Hospital, AP-HP, Medical Oncology Department, Angiogenesis Inhibitors Multidisciplinary Study Group (CERIA), Paris, France E-mail: [email protected] 2 Paris Descartes University, Cochin Hospital, AP-HP, Pharmacokinetics and Pharmacochemistry Unit, Angiogenesis Inhibitors Multidisciplinary Study Group (CERIA), Paris, France

Expert Opin. Investig. Drugs (2015) 24(5)

15

Investigational therapies up to Phase II which target PDGF receptors: potential anti-cancer therapeutics.

The platelet-derived growth factor receptor (PDGFR) pathway has important functions in cell growth and, by overexpression or mutation, could also be a...
459KB Sizes 0 Downloads 7 Views