Curr Oncol Rep (2014) 16:379 DOI 10.1007/s11912-014-0379-z

NEURO-ONCOLOGY (M GILBERT, SECTION EDITOR)

Targeted Therapy in Gliomas Mohamed Ali Hamza & Mark Gilbert

# Springer Science+Business Media New York 2014

Abstract The survival outcome of patients with malignant gliomas is still poor, despite advances in surgical techniques, radiation therapy and the development of novel chemotherapeutic agents. The heterogeneity of molecular alterations in signaling pathways involved in the pathogenesis of these tumors contributes significantly to their resistance to treatment. Several molecular targets for therapy have been discovered over the last several years. Therapeutic agents targeting these signaling pathways may provide more effective treatments and may improve survival. This review summarizes the important molecular therapeutic targets and the outcome of published clinical trials involving targeted therapeutic agents in glioma patients. Keywords Malignant gliomas . Gliomas . Radiation therapy . Chemotherapeutic agents . Oncology . Neuro-oncology . Molecular targets . Glioma patients . Central nervous system (CNS) . Flioblastoma (GBM) . Anaplastic gliomas . Therapeutic agents

Malignant gliomas refer to WHO grade III (anaplastic gliomas) and WHO grade IV refer to glioblastoma (GBM) and gliosarcoma [1]. Treatment options of gliomas include surgery, radiotherapy and chemotherapy. Although there is a standard treatment for glioblastoma – the most common and most aggressive type of gliomas – that includes radiotherapy with concurrent temozolomide followed by adjuvant temozolomide [2], there is no established standard treatment for less aggressive (lower grade) gliomas. In addition, the prognosis for glioblastoma continues to be poor. There is a desperate need for novel effective treatments for these devastating tumors. The accumulated wealth of knowledge about the molecular alterations involved in gliomagenesis and the increasing availability of novel molecularly targeted therapeutics may provide more effective treatment modalities. In this review, we discuss the most common alterations in the molecular pathways and the published clinical trials involving molecularly targeted therapies for gliomas.

Therapeutic Molecular Targets Introduction Cell Surface Molecular Targets Gliomas are the most frequent central nervous system (CNS) primary tumors, constituting more than 50 % of all primary CNS tumors. The World Health Organization (WHO) classified gliomas into four grades according to histopathology.

This article is part of the Topical Collection on Neuro-oncology M. A. Hamza (*) : M. Gilbert The University of Texas MD Anderson Cancer Center, Houston, TX, USA e-mail: [email protected] M. Gilbert e-mail: [email protected]

Epidermal Growth Factor Receptor (EGFR) EGFR is a member of the ErbB family, which consists of four receptors: HER1/EGFR, HER2, HER3 and HER4 [3]. EGFR is a 170 –kDa receptor tyrosine kinase that is formed of three domains: the extracellular domain, the transmembrane domain and the intracellular domain that contains the tyrosine kinase. Phosphorylation of the tyrosine kinase domain activates several signaling pathways, such as phosphatidylinositol 3’-kinase (PI3K)/AKT/mTOR, and Ras/mitogenactivated protein kinase (MAPK) [4]. Approximately 50 % of GBMs have amplification of EGFR, and overexpression of

379, Page 2 of 14

EGFR is common in malignant gliomas independent of amplification status [5]. The mutant receptor EGFRvIII has a deletion of exons 2-7 that causes a defect in the extracellular ligand-binding domain that results in constitutive activation of the receptor [6]. In addition, EGFRvIII may be an independent prognostic factor for poor survival outcome [7, 8]. Activation of EGFR pathways results in biological processes contributing to gliomagenesis including cell proliferation, angiogenesis, migration and survival. In addition, EGFR overexpression in GBM is associated with resistance to radiation therapy [9]. Platelet-Derived Growth Factor Receptor (PDGFR) The PDGF family signal through the alpha and beta PDGF receptor tyrosine kinases. The binding of the ligands to the extracellular domain of the receptor induces dimerization and cross-tyrosine phosphorylation of the intracellular domains. This results in the activation of several pathways including MAPK, PI3K, Jak family kinase, Src family kinase and phospholipase C-gamma. Approximately one- third of glioblastoma show overexpression of PDGFR-alpha [10]. Overexpression of PDGFR may be an important alteration in the transformation of low-grade gliomas to malignant gliomas [11]. In addition, analyses by the Cancer Genome Atlas Research Network showed that the proneural subtype of glioblastoma is characterized by aberrations of the PDGFR-alpha pathway, in addition to isocitrate dehydogenase-1 (IDH1) mutations [12]. Moreover, PDGFRs are involved in angiogenesis [13]. Vascular Endothelial Growth Factor (VEGF) VEGF is an important factor involved in neoangiogenesis which a characteristic of GBM [14]. Expression of VEGF is stimulated by hypoxia, acidosis, and growth factors such as PDGF, EGF, c-kit and hepatocyte growth factor (HGF). Binding of VEGF to its receptors VEGFR-1 and VEGFR-2 leads to phosphorylation of tyrosine kinase and activation of downstream signaling pathway including PI3K/Akt/PBK and Ras/MAPK [15]. The effects of VEGF include endothelial cell proliferation, cell migration, extracellular matrix degradation, and expression of proangiogenic factors including plasminogen activator inhibitor-1, urokinase-type plasminogen activator and matrix metalloproteinase-1 [16]. Integrins Integrins are heterodimeric cell surface adhesion receptors that function as receptors for extracellular matrix proteins including laminins, collagens, fibronectin, and vitronectin [17]. Binding of the ligand to the extracellular domain of the integrin receptor, which consists of two non-covalently associated alpha and beta subunits, results in activation of a

Curr Oncol Rep (2014) 16:379

signaling pathway involved in proliferation, migration, cell adhesions, differentiation and survival [18]. The integrins αvβ3 and αvβ5 are involved in the angiogenesis and tumor progression and are expressed in multiple malignancies including gliomas [19]. Intracellular Effectors/Pathways PI3K/AKT/mTOR Pathways PI3K is a serine / threonine kinase the activation of which results in phosphorylation and activation of downstream effectors including Akt and PBK that regulate cell growth, proliferation and apoptosis [20]. PI3K is activated by several receptor tyrosine kinases, integrins, and activated Ras. Akt is activated in approximately 70 % of GBM, in association with loss of PTEN (which leads to constitutive activation of the PI3K pathway) and/or activation of EGFR and PDFR tyrosine kinases [21]. Alteration of PTEN expression is present in 2040 % of GBM and is associated with a worse prognosis, and a mammalian target of rapamycin (mTOR) was identified as a critical downstream effector in PTEN/Akt signaling [22]. In addition, mTOR is also activated by Ras pathway, and activated mTOR activates ribosomal S6 kinase and inhibits the eukaryotic initiation factor 4E-binding protein-1, which is required for progression from G1 to S phase [7]. Protein Kinase C (PKC) Astrocytoma cells overexpress PKC, and the level of PKC activity was found to correlate with proliferation of astrocytoma cells [23]. PKC is involved in the downstream signaling of several growth factors that stimulates glioma cell proliferation including PDGF and EGF [24]. Activated PKC induces phosphorylation and activation of other downstream signaling effectors including Ras, Raf and MAPK [25]. Mitogen-Activated Protein Kinase (MAPK) Pathway The MAPK pathway involves activation of several protein kinases including Ras, Raf, Mek and Erk. Activation of Ras, a small guanine triphosphate-binding protein, results in activation of Raf, a serine/threonine kinase, which phosphorylates and activates MEK, and MEK subsequently phosphorylates and activates MAPK (also known as ERK) [26, 27]. These kinases activate downstream effectors that regulate transcription, translation and cytoskeletal rearrangement. Gliomas often have increased Ras activity as a result of mutation or amplification that result in activation of upstream growth factor receptors [28]. Farnesylation was found to be the rate–limiting step in Ras maturation [29]. Activation of Erk was found to correlate with poor outcome in GBM patients [30].

Curr Oncol Rep (2014) 16:379

Src and Src-Family Kinases (SFKs) Src and SFKs are effectors in multiple signaling pathways that regulate different aspects of tumor invasion and metastasis including proliferation, cell growth, adhesion, migration and invasion. Src and FYN (an SFK) were found to mediate EGFR and EGFRvIII signaling in a rodent GBM model [31]. Src inhibition decreases GBM cell viability and migration in vitro and decreased growth in vivo [32]. Src and SFKs activation and SFKs overexpression were reported in GBM cell lines and GBM patients [31]. Hepatocyte Growth Factor Receptor (c-MET) c-MET is a receptor tyrosine kinase for the hepatocyte growth factor, and aberrant signaling by the MET receptors and its potential role in tumor genesis has been reported [33]. In a study of 62 glioblastoma samples, c-MET overexpression was detected in 29 % of patients, and was associated with shorter overall survival when compared to those patients with little or no c-MET expression [34]. Other Molecular Targets

Page 3 of 14, 379

experimental approach for treatment of malignant glioma [40]. Death-receptor ligand activation may have an inhibiting effect on invasion, as demonstrated using anti-CD95 antibody treatment of mouse glioblastoma models [41]. Heat-Shock Proteins (HSPs) HSPs are protein chaperones that are overexpressed in several malignancies including gliomas [42]. HSPs are involved in the stability and activity of several proteins that stimulate growth, inhibit apoptosis, and promoting cell survival [43]. Inhibitors of HSPs may have a potential effect on potentiating signaling modulators and response to chemotherapy in gliomas [44]. Poly [ADP-Ribose] Polymerase (PARP) PARP is a DNA repair enzyme that is highly expressed in several types of cancer, and is thought to be implicated in the resistance of tumors to DNA damaging anticancer agents and radiotherapy [45]. PARP-1 is found to be expressed in the nuclei of GBM cells but not in normal brain tissue [46]. PARP inhibitors, in combination with DNA damaging agents may have a potential role in the treatment of gliomas.

Histone Deacetylases Nucleosomes are units of DNA organized by histone proteins. The acetylation and deacetylation (by histone deacetylases or HDACs) of the histone proteins may play an important role in the epigenetic modification of gene expression [35]. HDACs cause deacetylation of histones and condensation of the nucleosomes, which restricts access to the DNA [36]. Malignant gliomas have altered histone processing, and it is possible that HDAC inhibitors may increase the expression of certain genes that prevent or slow growth of tumors through mechanisms including induction of differentiation, cell cycle arrest and apoptosis [36, 37]. The Proteasome The proteolytic cellular functions of the proteasome include the cell cycle, inflammation, protein homeostasis and apoptosis [38]. Proteasome inhibitors may induce cell growth arrest and apoptosis and possibly decrease resistance to chemotherapy. Inhibition of proteasome activity may disrupt the degradation of proapoptotic proteins and enhance the degradation of NF-κB, which counteracts apoptosis [39]. Death Receptor Apoptosis can be induced through the activation of death receptors including Fas, TNFαR, DR3, DR4, and DR5 by their respective ligands. Death-receptor targeting has been an

Clinical Trials Involving Molecular Targeting Therapy for Gliomas Molecularly-targeted therapies are currently the focus of clinical research in primary brain tumors. The number of published clinical trials involving molecular-targeting agents has exceeded one hundred and is increasing (Tables 1 and 2). These publications started as early as twenty years ago with a single trial investigating the outcome of radiolabeled antiEGFR monoclonal antibodies in malignant astrocytoma [119]. The last several years have seen a dramatic increase in the number of trials with molecularly-targeted therapies for gliomas, with more than 20 trials of targeted agent-based clinical trials published in 2012. The most common molecular targets in early trials with single-target agents were inhibitors of EGFR and EGFRvIII, and inhibitors of VEGF and VEGFR. More recently, agents targeting different receptors such as integrins, or those inhibiting downstream signaling effectors such as Ras, MAPK and mTOR were studied more frequently in trials. However, with the exception of some trials with agents inhibiting EGFRvIII, molecular profile-based stratification of patients or analysis of outcome was not done in these trials. Single agent/single-target therapies were the most commonly investigated in glioma trials for many years; however, in the last few years this trend has reversed in favor of agents targeting multiple receptors or downstream effectors. In 2012, the ratio of the number of trials investigating multiple-target

Bevacizumab

Romidepsin

AMG 102 (rilotumumab) Trabedersen

VEGF

HDACs

HGF/SF

Cediranib

Cilengitide

anti-EGFR (125)I-mAb 425 Bortezomib

Enzastaurin

Enzastaurin

Integrins v3-v5

EGFR

Proteasome

PKC-beta

PKC-beta

Gefitinib

EGFR

VEGFR1, 2,3

Everolimus

Erlotinib

mTOR

EGFR

Tipifarnib

Temsirolimus

Farnesyl transferase

mTOR

TGF-β2

Bevacizumab

VEGF

Bevacizumab

Cilengitide

TLN-4601

Ras-MAPK

VEGF

Enzastaurin

Everolimus

mTOR

PKC-beta

Enzastaurin

PKC-beta

Integrins v3-v5

Vorinostat

Bevacizumab

Erlotinib

EGFR

HDACs

Dasatinib

SRC

VEGF

Everolimus

Gefitinib

mTOR

EGFR

Agents

Target

Enzastaurin

Enzastaurin, temozolomide, RT

Bortezomib

Cilengitide, temozolomide, RT anti-EGFR (125)I-mAb 425

Cediranib

Gefitinib

Erlotinib

Temsirolimus, temozolomide, RT Everolimus, temozolomide, RT

Tipifarnib

Trabedersen

AMG 102 (rilotumumab)

Bevacizumab, carboplatin, irinotecan Bevacizumab, temozolomide, RT, irinotecan Romidepsin

Cilengitide

Bevacizumab, carboplatin, irinotecan Enzastaurin, temozolomide

TLN-4601

Everolimus, temozolomide

Enzastaurin, temozolomide

Bevacizumab, irinotecan,

Vorinostat, temozolomide

Erlotinib

Dasatinib, CCNU

Gefitinib, RT

Everolimus, temozolomide, RT

Regimen

Table 1 Published clinical trials with single-target agents

I/II

I

I

II

I/II

II

II

II

I

I

I

IIb (RC)

II

I/II

II

II

II

II

II

II

I

I

II

I

I/II

I/II

I/II

I

Phase

Recurrent high-grade glioma

Newly diagnosed GBM

Recurrent malignant glioma

Newly diagnosed GBM

Newly diagnosed GBM

Recurrent GBM

Newly diagnosed GBM

Recurrent GBM (first relapse)

Newly diagnosed GBM

Newly diagnosed GBM

Recurrent/refractory high-grade glioma Newly diagnosed GBM

Recurrent GBM

Recurrent malignant glioma

Newly diagnosed GBM

Recurrent GBM (bev-resistant)

Recurrent GBM

Newly diagnosed GBM

Recurrent GBM (bev-naïve)

Newly diagnosed or recurrent GBM Newly diagnosed and recurrent GBM First recurrence of GBM

Recurrent primary brain tumors

High-grade gliomas

Recurrent/refractory GBM or AA

Recurrent GBM

Newly diagnosed GBM

Newly diagnosed GBM

Disease status

118

12

66

192

52

31

98

40

18

25

51

145

60

8/35

75

25

30

66

40

20

32

28

85

59

11

26

31/147

25

MTD 70 mg/week

[69]

[68]

[67]

[66]

DLTs thrombosis, thrombocytopenia, hemorrhage. RR 25 %, PFS6 7 % in GBM and 16 % in AG

MTD 2.5 mg/m2 in +EIASD and 1.7 mg/m2 in -EIASD patients. DLTs thrombocytopenia, MTD 250 mg daily

OS 15.7 months, 2-year OS 25.5 %

[74]

[73]

[72]

[71]

PFS6 69 %, PFS12 33 %, PFS 8 months, OS 16.1 months [70]

PFS6 25.8 %, RR 56.7 %.

PFS at 1 year 16 %, OS at 1 year 54 %

RR 6.3 %, PFS6 20 %, OS 9.7 months

[65]

[64]

[63]

Superior safety of trabedersen 10 μM when compared to trabedersen 80 μM and standard chemotherapy MTD 300 mg bid DLTs 25 % of grade 4/5 infections

[62]

[61]

[60]

[59]

[58]

[57]

[56]

[55]

[54]

[53]

[52]

[51]

[50]

[49]

[48]

[47]

Ref.

RR 0 %, PFS 4.3 weeks, OS 5.4 months

PFS 8 weeks, OS 34 weeks

PFS 14.2 months, OS 21.2 months

PFS6 16 %, OS 5.8 months

PFS6 12 %, drug detected in tumor specimen.

PFS 36 weeks, OS 74 weeks

PFS6 46.5 %, OS 8.3 months

PFS6 0 % , RR 0 %*

MTD everolimus 10 mg daily

MTD 500 mg daily

RR: GBM 25 %, AG 21 %, none in WHO grade II

DLTs: rash 90 %, diarrhea 60 %. PFS 1.9 months, OS 6.9 months MTD 500 mg daily

DLTs 38 %, PFS 1.3 months, PFS6 7.7 %

MTD 500 mg daily, OS 11.5 months

MTD 10 mg daily

No. of Outcome patients

379, Page 4 of 14 Curr Oncol Rep (2014) 16:379

Enzastaurin or lomustine

Erlotinib, temozolomide, RT

Bevacizumab

Cilengitide

Bevacizumab

Enzastaurin

Erlotinib

Bevacizumab

EGFRvIII-targeted dendritic cell-based vaccine Bevacizumab

Talampanel

Integrins v3-v5

VEGF

PKC-beta

EGFR

VEGF

EGFRvIII

Bortezomib, temozolomide, RT Erlotinib, temozolomide, RT Cilengitide Erlotinib, temozolomide, RT

Bevacizumab

Bortezomib

Erlotinib

Cilengitide

Erlotinib

Perillyl alcohol

VEGF

Proteasome

EGFR

Integrins v3-v5

EGFR

Ras-MAPK

Perillyl alcohol

Bevacizumab, irinotecan

Vorinostat

Lapatinib

Lapatinib

Vorinostat

HDACs

Bevacizumab: alone or plus irinotecan, Talampanel, temozolomide, RT

I/II

I/II

II (R)

II

I

II

II

I/II

II

II (R)

I

EGFRvIII-targeted dendritic cell-based vaccine

AMPA glutamatergic receptor EGFR/ErbB2

VEGF

II

II

III

II

II

II

II

II

Bevacizumab, etoposide

Bevacizumab

EGFRvIII-targeted peptide vaccine Bevacizumab, temozolomide, RT Bevacizumab, temozolomide (daily) Cilengitide, temozolomide, RT

VEGF

VEGF

I

II

Phase

Bevacizumab, temozolomide II or etoposide Bevacizumab, temozolomide, RT II

EGFRvIII-targeted peptide vaccine Bevacizumab

Bevacizumab

VEGF

Erlotinib

Bevacizumab

Erlotinib

EGFR

Erlotinib

VEGF

Erlotinib

EGFR

Regimen

EGFRvIII

Agents

Target

Table 1 (continued)

70

23

32

96

Recurrent malignant glioma

Newly diagnosed GBM

Recurrent GBM

Newly diagnosed and recurrent high-grade tumors Newly diagnosed GBM

Recurrent GBM

Recurrent GBM

Recurrent GBM

Newly diagnosed GBM

Recurrent GBM

Newly diagnosed GBM (EGFRvIII expressing)

Recurrent malignant glioma

Newly diagnosed GBM

Recurrent glioblastoma

Recurrent malignant glioma

Newly diagnosed GBM

Recurrent GBM

37

97

81

65

27

48

66

24

72

167

12

59

27

266

31

112

32

PFS6 was 48.2 % for GBM, 60 % for AA and 66.6 % for AO

PFS6 15 % and OS 9.9 months in patients received 2000 mg OS 15.3 months

OS 19.3 months

No DLTS with 1.3 mg/m2

PFS 16 weeks, PFS6 29 %, OS 31 weeks

PFS6 17.3 %, OS 5.7 months

RR 0 %, no DLTs with 1500 mg bid*

PFS6 42.6 % and 50.3 %,, RR 28.2 % and 37.8 %, and OS 9.2 months and 8.7 months, respectively OS 18.3 months, 24-month OS 41.7 %

PFS6, RR and OS were 44.4 % and 40.6 %, 37 % and 22 %, and 44.4 and 63.1 weeks for GBM and AG, respectively MTD not reached, PFS 6.8 months, OS 18.7 months

PFS6 33.9 %, PFS 3.3 months, OS10.5 months, 1-year OS 34.5 %, RR27.6 % PFS and OS were 1.5 months and 1.6 months, and 6.6 months and 7.1 months for Enzastaurin and lomustine, respectively PFS 2.8 months, OS 8.6 months*

OS 19.7 months

PFS6 85 %, PFS12 51 %, 12 and 24 months OS 85.1 % and 42.5 % PFS6 18.8 %, PFS 15.8 weeks, OS 37 weeks.

PFS6 67 %, OS 26 months

PFS 13.6 months, OS 19.6 months

PFS6 4.4 %, PFS 7.3 weeks

PFS6 and PFS in recurrent GBM 3 % and 2 months, in recurrent AG 27 % and 2 months, 12-month OS was 57 % in newly diagnosed GBM MTD 650 mg daily, DLT rash

No. of Outcome patients

Newly diagnosed GBM (EGFRvIII 21 expressing) Newly diagnosed GBM 51

Newly diagnosed GBM

Newly diagnosed GBM and recurrent malignant glioma and meningiomas Recurrent GBM (bev-resistant)

Newly diagnosed GBM and recurrent malignant glioma

Disease status

[97]

[96]

[95]

[94]

[93]

[92]

[91]

[90]

[89]

[88]

[87]

[86]

[85]

[84]

[83]

[82]

[81]

[80]

[79]

[78]

[77]

[76]

[75]

Ref.

Curr Oncol Rep (2014) 16:379 Page 5 of 14, 379

Gefitinib

Tipifarnib

Gefitinib

Tipifarnib

VEGF

Farnesyl transferase

EGFR

Farnesyl transferase

anti-EGFR (125)I-mAb 425

Recurrent malignant glioma

High-grade gliomas

Newly diagnosed GBM

Recurrent GBM

Recurrent high-grade glioma

Recurrent GBM (PTEN-deficient)

Newly diagnosed GBM

Recurrent malignant glioma

Recurrent GBM

Newly diagnosed GBM

Disease status

II

I/II

I/II

II

II

I

II

II

I

I/II

I

Newly diagnosed high-grade astrocytoma Malignant astrocytoma

Recurrent malignant glioma

Recurrent and progressive GBM

Recurrent GBM

Malignant glioma

Recurrent GBM

Recurrent GBM

Newly diagnosed high-grade glioma Stable or progressive malignant glioma

Newly diagnosed GBM

III (RCD) Newly diagnosed GBM

II

II

I

II

I

I

II

I

II

II

Phase

25

59

16

44

57

41

65

43

83

29

20

162

89

28

13

35

11

14

10

25

43

28

1-year OS 60 %, OS 15.6 months

No DLTs, 1-year OS 58 %, OS 13.5 months

PFS6 39 %, PFS 17 weeks, OS 45 weeks, 12-month PFS 16 % MTD 200 mg 3 times /week for 4 weeks. PFS3 0 %

RR 19.5 %, RR 44.4 % in patients with low level of phosphorylated PKB/Akt PFS6 13 %, PFS 8.1 weeks, OS 39.4 weeks

RR 36 %, PFS6 7.8 %, PFS 2.3 months, OS 4.4 months

PFS6: 9.1 % in AG, 11.9 % in GBM, 16.7 % in GBM not receiving EIAED OS 37.9 weeks for placebo and 42.9 weeks for marimastat MTD not reached at 150 mg daily in non-EIAED patients and 200 mg daily in EIAED patients. PFS 26 weeks, OS 55 weeks No DLTs, RR 37.9 %, OS 17.7 months in GBM, OS not reached in AG MTD: erlotinib alone 200 mg daily for non-EIAED patient and 500 mg daily for EIAED patients. Erlotinib plus temozolomide 450 mg daily for EIAED patients. PFS6 10.5 % PFS6 3.5 %, PFS 9 weeks

PFS6 14.3 %, PFS 8.4 weeks, OS 24.6 weeks

MTD 200 mg daily, OS 12 months

MTD 3 mg of nimotuzumab labeled with 10 mCi of (188)Re PFS6 46 %, 6-month OS 77 %.

Tumor cell proliferation decreased in 50 % of patients

Toxicity: DVT, PE.

MTD 70 mg daily, OS 6 months

PFS 9 weeks, PFS6 14 %, OS 30 weeks

OS 7.7 months*

No. of Outcome patients

[119]

[118]

[117]

[116]

[115]

[114]

[113]

[112]

[111]

[110]

[109]

[108]

[107]

[106]

[105]

[104]

[103]

[102]

[101]

[100]

[99]

[98]

Ref.

*Early termination

AA: anaplastic astrocytoma, AG: anaplastic glioma, GBM: glioblastoma, RR:response rate, EIAED: enzyme-inducing anti-epileptic drugs, MTD: maximum tolerated dose, DLT: dose-limiting toxicity, RR:response rate, PFS: progression free survival, PFS6: 6-month progression free survival, PFS12:12-month progression free survival, OS: overall survival, R: randomized, RC: randomized and placebocontrolled, RCD: randomized, placebo-controlled and double-blind

EGFR

anti-EGFR (125)I-mAb 425

EMD 55,900

anti-EGFR (125)I-mAb 425 anti-EGFR (125)I-mAb 425

EMD 55,900

EGFR

Marimastat, temozolomide

Gefitinib

Marimastat

Erlotinib alone or with temozolomide Gefitinib

EGFR

Erlotinib

EGFR

Temsirolimus

Temsirolimus

Erlotinib alone or with temozolomide

Matrix metallo-protease EGFR

Temsirolimus

Temsirolimus

Erlotinib

EGFR

mTOR

Nimotuzumab

EGFR

mTOR

Erlotinib, RT

Erlotinib

EGFR

Nimotuzumab, RT

Marimastat

Matrix metallopr-otease Marimastat

Tipifarnib

Tipifarnib, RT

Rapamycin

Bevacizumab, temozolomide, RT Rapamycin

Nimotuzumab Nimotuzumab labeled with labeled with 188 Re 188 Re Bevacizumab Bevacizumab, irinotecan

Bevacizumab

VEGF

Atrasentan

EGFR

Atrasentan

Endothelin-A receptor

Erlotinib, carboplatin

Tipifarnib (neoadjuvant)

Regimen

mTOR

Tipifarnib

Erlotinib

Farnesyl transferase

EGFR

Agents

Target

Table 1 (continued)

379, Page 6 of 14 Curr Oncol Rep (2014) 16:379

Sorafenib, temsirolimus Sunitinib Thalidomide, RT

Bevacizumab, erlotinib

Vandetanib

Sorafenib, temsirolimus

Sunitinib

VEGF, EGFR

VEGFR2/EGFR

II II

Erlotinib, Sorafenib Nintedanib

Sunitinib Bevacizumab, everolimus, temozlolmide, RT AEE788 Vorinostat, bortezomib Thalidomide, irinotecan

Sunitinib

Bevacizumab, everolimus

AEE788

Vorinostat, bortezomib

VEGFR/PDGFR/ c-kit/FLT3 VEGF, mTOR

EGFR/ErbB/VEGFR2

HDACs, proteasome

Cetuximab, bevacizumab

EGFR, VEGF

Pazopanib

Vandetanib

Vatalanib

VEGFR/c-kit/ PDGFR

VEGFR2/EGFR

VEGFR/PDGFR/c-kit

VEGF, bFGF, HGF, IL-8 ABT-510

Bevacizumab, Erlotinib

Sorafenib

Vatalanib

Thalidomide

VEGF/bFGF

VEGF, EGFR

Vatalanib, temozolomide, RT

Aflibercept

RAF/VEGFR/PDGFR

Aflibercept

Sunitinib

VEGFR/PDGFR/ c-kit/FLT3 VEGF, placental growth factor VEGFR/PDGFR/c-kit

Vandetanib, temozolomide, RT Vatalanib, temozolomide, RT

Pazopanib

Cetuximab, bevacizumab, irinotecan ABT-510, temozolomide, RT

Sorafenib, TMZ

Thalidomide, temozolomide, isotretinoin,celecoxib Bevacizumab, Erlotinib

Vorinostat, bevacizumab, irinotecan Sunitinib, irinotecan

Thalidomide

Vorinostat, bevacizumab

VEGF/bFGF

HDACs, VEGF

Erlotinib, sirolimus

Thalidomide

Erlotinib, sirolimus

EGFR, mTOR

I/II

I

II

I

II

II

II

I

I

II

I

I

II

II

I

II

II

I

II

II

I/II

Bevacizumab or bevacizumab II plus Erlotinib Vandetanib I/II

I/II

II

II

34/41

13

43

Recurrent GBM

25

89

63

13/18

15/64

10

19

39

37

64

68

19 54

Newly diagnosed GBM

Newly diagnosed GBM

Recurrent GBM

Newly diagnosed GBM

Recurrent GBM

Newly diagnosed GBM

20

13

35

23

43

47

Recurrent malignant glioma 57

Newly diagnosed GBM

Newly diagnosed GBM

Recurrent malignant glioma 58

Recurrent malignant glioma 25

Recurrent GBM

Recurrent AG

Recurrent GBM

Recurrent GBM

Newly diagnosed GBM

Recurrent malignant gliomas 30

Recurrent malignant gliomas 19

Newly diagnosed GBM

Recurrent GBM

Recurrent GBM

Recurrent AG/GBM

Recurrent GBM

[139]

[138]

[137]

[136]

[135]

[134]

[133]

[132]

[131]

[130]

[129]

[128]

[127]

[126]

[125]

[124]

[123]

[122]

[121]

[120]

Ref.

[143]

[142]

[145]

MTD 1000 mg daily*

[147]

MTD 100 mg daily, DLTs GI bleeding, thrombocytopenia, neutropenia [146]

PFS 12 weeks, PFS6 3 %, OS 35 weeks

MTD not reached with 200 mg daily, PFS 45.9 weeks, OS 64.4 weeks. [144]

RR 34 %, PFS6 30 %, OS 29 weeks

PFS 6 months, 1-year PFS 16 %, OS 12 months

PFS6 and OS: GBM 28 % and 42 weeks, and AG 44 % and 71 weeks [141]

DLTs fatigue, rash, neutropenia. OS 20 months, 2-year survival 40 %. [140]

PFS6 GBM 7.7 % and AG 25 %. RR 25 %. PFS GBM12 weeks, AG 24 weeks MTD not reached

MTD sunitinib 50 mg daily, PFS6 24 %, RR4%

MTD vorinostat 400 mg days 1-7 and 15-21.

PFS6 36 %, PFS 13 weeks, OS 62 weeks

PFS6 0 %, PFS 1.5 months, OS 3.2 months*

DLTs proteinuria, diarrhea, hepatotoxicity; RR 17 %*

PFS6 and OS were 21.5 % and 12.1 months for AA and 16.7 % and 12.6 months for GB PFS 11.3 months, OS 13.9 months

MTD erlotinib 150 mg daily, sirolimus 5 mg daily

MTD 400 mg daily, PFS6 and OS were 6.5 % and 6.3 months in GBM and 7 % and 7.6 months in AG MTD sorafenib 800 mg daily and temsirolimus 25 mg weekly. PFS6 0 %, PFS 8 weeks* PFS6 and OS were 10.4 % and 9.4 months in the bev-naïve and 0 % and 4.4 months in the bev-resistant Median survival 10 months

RR 70 %, 6-mo nth PFS 70 %, PFS 8 months, OS 9.5 months

Stable disease 12 %, PFS 1 month, OS 6 months*

OS 5.7 months, 6-month PFS 14 %

RR 5 % , MTD not reached*

RR 0 %, PFS 8 weeks, OS 15 weeks*

6-month PFS 26 %, OS 7,4 months*

No. of Outcome patients

Recurrent/progressive GBM 18

Recurrent GBM

Recurrent GBM

Recurrent GBM

Phase Disease status

Pazopanib, Lapatinib

Temsirolimus, Bevacizumab

RAF/VEGFR/ PDGFR, mTOR VEGFR/PDGFR/ c-kit/FLT3 VEGF/bFGF

VEGFR/c-kit/ PDGFR, EGFR/ErbB EGFR, RAF/VEGFR/ Erlotinib, Sorafenib PDGFR VEGFR/FGFR/ PDGFR Nintedanib

Temsirolimus, bevacizumab Pazopanib, Lapatinib

Sorafenib, TMZ

Sorafenib

RAF/VEGFR/PDGFR

mTOR, VEGF

Regimen

Agents

Targets

Table 2 Published clinical trials for multiple-targets agents

Curr Oncol Rep (2014) 16:379 Page 7 of 14, 379

Imatinib, hydroxyurea

Imatinib

Imatinib

Imatinib

PDGFR/ c-kit/bcr-abl

PDGFR/ c-kit/bcr-abl

PDGFR/ c-kit/bcr-abl

Thalidomide

Imatinib

Thalidomide

Imatinib

Gefitinib, sirolimus

Imatinib

Thalidomide

Thalidomide

VEGF/bFGF

PDGFR/ c-kit/bcr-abl

VEGF/bFGF

PDGFR/ c-kit/bcr-abl

EGFR, mTOR

PDGFR/ c-kit/bcr-abl

VEGF/bFGF

VEGF/bFGF

Thalidomide

Thalidomide

Thalidomide , carmustine

Thalidomide, temozolomide, RT Thalidomide alone or with temozolomide Thalidomide

Imatinib, hydroxyurea

Gefitinib, sirolimus

Imatinib

Thalidomide, temozolomide

Thalidomide, cisplatin, temozolomide Imatinib, hydroxyurea

Lenalidomide

Lenalidomide, RT

Thalidomide, irinotecan

II

II

II

II

II

II

II

I

I/II

II

II

I

I

II

I

II

I

II

II

I

II

III

II

II

32

20

231

44

39

17

36

32

65

50

23

26

22

18

44

67

33

42 Recurrent high-grade glioma 39

Recurrent GBM

Recurrent high-grade glioma 40

Recurrent GBM

Newly diagnosed GBM

Newly diagnosed GBM

Recurrent GBM

Recurrent malignant glioma 34

Recurrent malignant glioma 50/55

Recurrent GBM

Recurrent AG

Malignant brain tumors

Recurrent brain tumors

Newly diagnosed malignant glioma Recurrent GBM

Newly diagnosed GBM

Newly diagnosed or recurrent GBM Newly diagnosed GBM

Recurrent GBM

[151]

[150]

[149]

[148]

Ref.

RR 6 %, 1-year OS 20.5 %

OS 31 weeks, 1-year OS 35 %

PFS 100 days, RR 24 %

OS and PFS 103 weeks and 36 weeks for thalidomide and temozolomide, and 63 weeks and 17 weeks for thalidomide. PFS 25 weeks, OS 36 weeks

PFS 22 weeks, OS 73 weeks

No DLTs with 1200 mg daily in non-EIAED patients. PFS6 3 % for GBM and 10 % in AG MTD: non- EIAED patients gefitinib 500 mg and sirolimus 5 mg daily, EIAED patients gefitinib 1000 mg and sirolimus 10 mg daily. DLTs: stomatitis, diarrhea, rash, thrombocytopenia. PFS6 27 %, PFS 14.4 weeks

RR 7 %, PFS6 24 %, PFS 15 weeks

PFS6 24 %, RR 10 %,

MTD 150 mg daily, DLTs thrombocytopenia and febrile neutropenia

MTD 20 mg/m2, PFS

Targeted therapy in gliomas.

The survival outcome of patients with malignant gliomas is still poor, despite advances in surgical techniques, radiation therapy and the development ...
337KB Sizes 2 Downloads 0 Views