Signal Transduction and Targeted Therapy for Gynecologic Cancer



Fig. 3.1
Targeting the angiogenic cascades in gynecologic cancer. Angiogenesis is regulated by a number of growth factor receptor pathways. The specific ligands bind to their receptors, and each tyrosine kinase activates the intracellular signaling cascade, including mitogen-activated protein kinase (MAPK) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt pathways. Subsequently, the pro-angiogenic signaling pathways are activated. VEGF vascular endothelium growth factor, VEGFR VEGF receptor, PlGF placental growth factor, FLT fms-related tyrosine kinase, KDR kinase insert domain receptor, FGF fibroblast growth factors, FGFR FGF receptor, PDGF platelet-derived growth factor, PDGFR PDGF receptor, Ang angiopoietin, Tie Tyrosine kinase with immunoglobulin-like and EGF-like domains



These VEGF ligands and PlGF uniquely bind to three structurally similar receptors: VEGFR1 [or fms-related tyrosine kinase 1 (FLT1)], VEGRF2 (or kinase insert domain receptor), and VEGFR3 (or FLT4). VEGF-A binds both VEGFR1 and VEGFR2, which are expressed mainly on vascular endothelial cells; VEFGR2 is predominant and mediates the angiogenic and vascular permeability effects of VEGF [4]. VEGF3 has been reported to play an important role in lymphangiogenesis through preferential binding to VEGF-C and VEGF-D. Neuropilin (NP)1 and NP2 (NRP1 and NRP2, respectively) act as VEGFR co-receptors, thus increasing the binding affinities of VEGFs to their receptors. Ligand binding activates multiple intracellular signaling cascades downstream of VEGFRs, including mitogen-activated protein kinase (MAPK), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt, phospholipase Cγ, and small GTPase pathways [5] and induces proangiogenic effects such as endothelial cell proliferation, migration, survival, and differentiation. VEGF also increases vascular permeability and vasodilation, causing interstitial hypertension and leaky neovasculature.

VEGF and VEGFR overexpression is observed in many solid tumors, including ovarian cancer, and has been associated with an increased risk of metastatic disease and poor prognosis, [68]. In ovarian cancer, higher levels of VEGF-A expression were observed in tumors from patients with platinum-resistant disease vs. those with platinum-sensitive disease [9]. VEGF-A and VEGFR2 coexpression has been detected in both ovarian cancer cells and ovarian tumor tissues, suggesting excision of the autocrine VEGF-A–VEFGR2 loop in ovarian cancer [10, 11]. A recent study found that increased Zeste homolog 2 (EZH2) expression in ovarian tumor cells or tumor vasculature was predictive of a poor clinical outcome [12], and VEGF-A stimulation, which promotes angiogenesis by methylating and silencing vasohibin1, directly led to an increase in endothelial EZH2 expression. These observations indicate that VEGF signaling pathways are promising therapeutic targets in ovarian cancer.


3.2.1.1 Bevacizumab


Bevacizumab is an intravenously (i.v.) administered recombinant humanized monoclonal IgG1 antibody that targets VEGF-A, with clinical benefits in patients with metastatic colorectal cancer, non-small cell lung cancer, and breast cancer [13]. This drug binds and neutralizes all biologically active forms of VEGF-A (e.g., VEGF-A165), thus suppressing tumor growth and inhibiting metastatic disease progression by inhibiting neovascularization and inducing existing microvessel regression [14, 15]. Bevacizumab also normalizes tumor vessels that are structurally and functionally abnormal. These morphological changes lead to functional changes (e.g., decreased interstitial fluid pressure, increased tumor oxygenation, improved drug penetration in tumors) that may enhance the effects of chemotherapy [16].

The phase II trials Gynecologic Oncology Group (GOG)-0170D and AVF2949 evaluated bevacizumab as a monotherapy for recurrent ovarian cancer and yielded favorable results, with response rates of 16–21% [17, 18] and hypertension and proteinuria as the most common grade 3/4 adverse events. Although no gastrointestinal (GI) perforation was observed in patients of the GOG-0170D study who had received one or two previous regimens, the AVF2949 trial observed GI perforation in five patients (11.4%) previously subjected to heavy treatment (three or more prior regimens).

Two landmark phase III trials of bevacizumab for ovarian cancer, GOG-0218 and International Collaborative Ovarian Neoplasm (ICON) 7, were conducted in a first-line/adjuvant chemotherapy setting (Table 3.1) [19, 20]. In the GOG-0218 trial, patients who received combination chemotherapy (paclitaxel/carboplatin) plus bevacizumab (15 mg/kg) for six cycles and maintenance bevacizumab for 16 cycles had a significantly longer progression-free survival (PFS) than those who received first-line chemotherapy alone (median PFS: 10.3 vs. 14.1 months) [19]. However, no statistically significant difference was observed in overall survival (OS). Similarly, patients in the ICON 7 trial were randomized to chemotherapy alone (carboplatin/paclitaxel) or plus bevacizumab (7.5 mg/kg) for six cycles, with 12 cycles of maintenance bevacizumab [20]. The latest report observed a significantly prolonged restricted mean survival time among poor-prognosis patients in the bevacizumab group vs. the chemotherapy group (39.3 vs. 34.5 months), although no OS benefit of bevacizumab was recorded [21].


Table 3.1
Phase III trials of targeted therapy in ovarian cancer







































































































































































































































































































































































Trial

Patients

Treatment

Median PFS (M)

Median OS (M)

Selected Adverse Events a

Anti-angiogenic agents

First-line treatment

GOG-0218 [19]

1873
       

Arm 1

625

CP + placebo → placebo

10.3

39.3

1.2% GI events (G ≥ 2), 7.2% HT (G ≥ 2), 5.8% VTE (any grade), 0.8% bleeding (G ≥ 3)

Arm 2

625

CP + Bevacizumab → placebo

11.2

38.7

2.8% GI events (G ≥ 2), 16.5% HT (G ≥ 2), 5.3% VTE (any grade), 1.3% bleeding (G ≥ 3)

Arm 3

623

CP + Bevacizumab → Bevacizumab

14.1 ***

39.7

2.6% GI events (G ≥ 2), 22.9% HT (G ≥ 2), 6.7% VTE (any grade), 2.1% bleeding (G ≥ 3)

ICON7 [20]

1528
       

All patients

Arm 1

764

CP

17.5

58.6

1.3% GI events, 0.3% HT, 1.7% VTE, 0.3% bleeding

Arm 2

764

CP + Bevacizumab → Bevacizumab

19.9

58.0

2.1% GI events, 6.2% HT, 4.3% VTE, 1.2% bleeding

High-risk patients

Arm 1

254

CP

10.5

30.2
 

Arm 2

248

CP + Bevacizumab → Bevacizumab

16.0 *

39.7 *
 

GOG-0262 [22]

692
       

All Patients

Arm 1

346

CP ± Bevacizumab → Bevacizumab

14.0

39.0

15.7% anemia, 83.4% neutropenia

Arm 2

346

Weekly CP ± Bevacizumab → Bevacizumab

14.7

40.2

36.5% anemia, 72.4% neutropenia

Bevacizumab (+)

Arm 1

289

CP + Bevacizumab → Bevacizumab

14.7

 

Arm 2

291

Weekly CP + Bevacizumab → Bevacizumab

14.9

 

Bevacizumab (−)

Arm 1

57

CP

10.3

 

Arm 2

55

Weekly CP

14.2 *

 

AGO-OVAR12 [40]

1366
       

Arm 1

455

CP + placebo → placebo

16.6


2.0% diarrhea, 0.4% HT, 6.9% anemia, 6.4% thrombocytopenia, 36.0% neutropenia

Arm 2

911

CP + Nintedanib → Nintedanib

17.2 *


21.5% diarrhea, 4.7% HT, 13.5% anemia, 17.7% thrombocytopenia, 42.1% neutropenia

Maintenance

AGO-OVAR16 [37]

940
   
2nd interim OS analysis
 

Arm 1

468

Pt CT → placebo

12.3

HR = 1.08 (0.87–1.33)

5.6% HT, 1.5% neutropenia, 0.7% liver-related toxicity, 1.1% diarrhea, 0.2% fatigue, 0.7% thrombocytopenia, 0.2% palmar-plantar erythrodysesthesia

Arm 2

472

Pt CT → Pazopanib

17.9 *

30.8% HT, 9.9% neutropenia, 9.4% liver-related toxicity, 8.2% diarrhea, 2.7% fatigue, 2.5% thrombocytopenia, 1.9% palmar-plantar erythrodysesthesia

Recurrent disease

OCEANS [23, 25]

484
       

Arm 1

242

CG + placebo

8.4

35.2

0.4% HT, 0.9% Proteinuria, 2.6% VTE

Arm 2

242

CG + Bevacizumab

12.4 ***

33.3

17.4% HT, 8.5% Proteinuria, 4.0 VTE

AURELIA [24]

361
       

Arm 1

182

chemotherapy alone b

3.4

13.3

1.1% HT, 4.4% TEE

Arm 2

179

chemotherapy + Bevacizumab

6.7 **

16.6

7.3% HT, 5.0% TEE, 1.7% proteinuria, 1.7% GI perforation

ICON6 [34]

456
   
OS data immature
 

Arm 1

118

Pt CT + placebo → placebo

8.7

21.0

[n = 115] 7.8% fatigue, 3.5% HT, 1.7% diarrhea, 6.1% nausea/vomiting, 3.5% febrile neutropenia, 23.5% neutropenia, 2.6% thrombocytopenia

Arm 2

174

Pt CT + Cediranib → placebo

9.9


[n = 329] 16.4% fatigue, 11.6% HT, 10.3% diarrhea, 7.0% nausea/vomiting, 6.7% febrile neutropenia, 25.6% neutropenia, 7.6% thrombocytopenia (during chemotherapy phase: Arm 2/3)

Arm 3

164

Pt CT + Cediranib → Cediranib

11.0 *

26.3

TRINOVA-1 [51]

919
       

Arm 1

458

Weekly PTX + placebo

5.4

17.3

Any grade: 25.7% edema, 0.2% GI perforation, 3.5% HT, 3.8% VTE, 16.6% bleeding

Arm 2

461

Weekly PTX + Trebananib

7.2 ***

19.0

Any grade: 57.3% edema, 1.5% GI perforation, 6.1% HT, 6.3% VTE, 10.0% bleeding

EGFR inhibitors

Maintenance

EORTC 55041 [69]

835
       

Arm 1

415

Pt CT → observation

12.4

59.1
 

Arm 2

420

Pt CT → Erlotinib

12.8

50.8

12.8% rash, 4.8% diarrhea

FRα inhibitors

Recurrent disease

Farletuzumab [89]

1091
       

Arm 1

352

PtTx CT + placebo → placebo

9.0

29.1

41.2% neutropenia, 8.0% thrombocytopenia, 13.6% leukopenia, 9.9% anemia

Arm 2

376

PtTx CT + Farletuzumab (1.25 mg/kg) → Farletuzumab

9.5

28.7

44.4% neutropenia, 13.0% thrombocytopenia, 11.7% leukopenia, 10.1% anemia

Arm 3

363

PtTx CT + Farletuzumab (2.5 mg/kg) → Farletuzumab

9.7

32.1

38.3% neutropenia, 11.6% thrombocytopenia, 9.9% leukopenia, 10.2% anemia

CA125 < 3 × ULN c

Arm 1

118

PtTx CT + placebo → placebo

8.8

29.1
 

Arm 2

174

PtTx CT + Farletuzumab (1.25 mg/kg) → Farletuzumab

9.5

NE
 

Arm 3

164

PtTx CT + Farletuzumab (2.5 mg/kg) → Farletuzumab

13.6 *

NE *
 


PFS progression-free survival, M months, OS overall survival, GOG Gynecologic Oncology Group, ICON International Co-operative Group for Ovarian Neoplasia, AGO Arbeitsgemeinschaft Gynäkologische Onkologie, EORTC European Organization for Research, EGFR epidermal growth factor receptor, FR folate receptor, CP carboplatin plus paclitaxel, Weekly CP weekly paclitaxel plus every 3-weeks carboplatin, CG carboplatin plus gemcitabine, PTX paclitaxel, Pt CT platinum-based chemotherapy, PtTx CT platinum- and taxane-based chemotherapy, HR hazard ratio, NE not estimated, GI gastrointestinal, G grade, HT hypertension, VTE venous thrombosis, TEE thromboembolic events

aSelected adverse events (grade ≥ 3), except for indicated

bInvestigator selected chemotherapy (pegylated liposomal doxorubicin, topotecan, or weekly paclitaxel)

cSubgroup with CA125 levels not more than three times the upper limit of normal (ULN)

* P < 0.05, ** P < 0.001, *** P < 0.0001 vs. control arm

A randomized trial, GOG-0262, evaluated the optimal combination of bevacizumab with dose-dense therapy (weekly paclitaxel plus carboplatin every 3 weeks) and conventional dose therapy (paclitaxel/carboplatin every 3 weeks; Table 3.1) [22]. Both groups of patients who opted to receive bevacizumab had a similar PFS, although among patients who did not receive bevacizumab, the medium PFS was 3.9 months longer with dose-dense therapy vs. conventional dose therapy (14.2 vs. 10.3 months).

Two phase III trials, OCEANS (Ovarian Cancer Study Comparing Efficacy and Safety of Chemotherapy and Anti- Angiogenic Therapy in Platinum-Sensitive Recurrent Disease) and AURELIA (Avastin Use in Platinum-Resistant Epithelial Ovarian Cancer), were conducted to evaluate recurrent disease (Table 3.1). Both trials evaluated the effect of bevacizumab in combination with chemotherapy and observed improvements in PFS [23, 24]. In the OCEANS study, platinum-sensitive recurrent ovarian cancer patients received six cycles of carboplatin/gemcitabine in combination with bevacizumab, followed by maintenance bevacizumab, and patients receiving bevacizumab had a significantly longer PFS vs. the control arm (median PFS: 12.4 vs. 8.4 months) [23]. However, the final OS analysis revealed no significant difference between the treatment arms [25]. In the AURELIA study, platinum-resistant recurrent ovarian cancer patients received single-agent chemotherapy [pegylated liposomal doxorubicin (PLD), weekly paclitaxel, or topotecan] alone or with bevacizumab. PFS was significantly improved in the chemotherapy plus bevacizumab arm vs. the chemotherapy arm (median PFS: 6.7 vs. 3.4 months). However, the trend toward improved OS (median: 16.6 vs. 13.3 months) was not statistically significant. GI perforation was observed only in the bevacizumab arm (2.2%), although the risk was lower than expected.

Several other ongoing phase III trials are investigating the optimal use of bevacizumab. The GOG-0252 study is evaluating the efficacy of bevacizumab in combination with intraperitoneal (i.p.) chemotherapy (i.v. paclitaxel and i.p. cisplatin or carboplatin) vs. i.v. chemotherapy (paclitaxel/carboplatin). The AGO-OVAR 17 (BOOST) trial is investigating the optimal bevacizumab treatment duration (15 vs. 30 months) with first-line chemotherapy (paclitaxel/carboplatin). GOG-0213, a study on platinum-sensitive recurrent disease, is comparing chemotherapy (paclitaxel/carboplatin) alone vs. with bevacizumab; surgical candidates in this cohort will undergo secondary randomization to surgery or no surgery.


3.2.1.2 Aflibercept


Aflibercept (VEGF Trap) is a fusion protein of the Fc region of immunoglobulin G1 with domain two of VEGFR1 and domain three of VEGFR2 (VEGFRδ1R2). This decoy receptor binds with high affinity to VEGF-A, thus preventing VEGFR1 and VEGFR2 binding and subsequent stimulation [26]. Aflibercept also exhibits a strong binding affinity for VEGF-B and PlGF.

Two phase II studies of platinum-resistant disease and symptomatic malignant ascites have been conducted (Table 3.2) [27, 28], and both demonstrated effective control of malignant ascites with aflibercept, evidenced by a reduction in the interval between repeat paracenteses (e.g., 55.1 vs. 23.3 days) [28]. However, one study observed a higher frequency of fatal GI events in the aflibercept arm (3/29 patients) vs. the placebo arm (1/26 patients) [28]. In the other study, platinum-resistant ovarian cancer patients were randomized to receive aflibercept at different doses (2 or 4 mg/kg) (Table 3.2) [29]. Although aflibercept was generally well tolerated at both doses, the response rate was low.


Table 3.2
Randomized phase II trials of targeted therapy in ovarian cancer










































































































































































































































































































































































































































































































































































































































































Agents

Patients (n) a

Treatment

Response Rate (%)

Median PFS (M)

Median OS (M)

Selected Grade ≥ 3 Adverse Events

Anti-angiogenic agents

Maintenance

Nintedanib [39]

83
   
36-week PFS
   

Arm 1

40

Chemotherapy → placebo


16.3%


8% hepatotoxicity

Arm 2

43

Chemotherapy → Nintedanib


5.0%


51% hepatotoxicity

Sorafenib [47]

246
         

Arm 1

123

PtTx CT → placebo


15.7


0.8% hand–foot skin reaction

Arm 2

123

PtTx CT → Sorafenib


12.7


39.0% hand–foot skin reaction, 14.6% rash

Recurrent disease
           

Aflibercept [27]

55
 
Time to repeat paracentesis
     

Arm 1

26

placebo

23.3 days



8% dyspnea, 44% fatigue/asthenia, 4% GI fistula

Arm 2

29

Aflibercept: 4 mg/kg every 2 weeks

55.1 days *



20% dyspnea, 13% fatigue/asthenia, 10% GI perforation, 8% proteinuria, 7% HT, 7% VTE

Aflibercept [28]

215
         

Arm 1

106

Aflibercept: 2 mg/kg every 2 weeks

0.9

13.0 weeks

59.0 weeks

25.5% HT, 9.4% proteinuria, 5.7% fatigue

Arm 2

109

Aflibercept: 4 mg/kg every 2 weeks

4.6

13.3 weeks

49.3 weeks

27.5% HT, 7.3% proteinuria, 3.7% fatigue

Pazopanib [38]

73
         

Arm 1

36

Weekly PTX

25

3.5

13.7

3% neutropenia, 6% fatigue, 3% leucopenia, 14% anemia

Arm 2

37

Weekly PTX + Pazopanib

56 *

6.5 **

19.1

30% neutropenia, 11% fatigue, 11% leucopenia, 8% hypertension, 8% raised aspartate aminotransferase or alanine aminotransferase, 5% anemia, 3% ileal perforation.

Sunitinib [43]

73
         

Arm 1

36

Sunitinib: 50 mg daily for 4 weeks in a 6-week cycle

17

4.8

13.6

4.5% increased γ-glutamyl transferase (% of all reported adverse events)

Arm 2

37

Sunitinib: 37.5 mg daily continuously

5

2.9

13.7

6.1% increased γ-glutamyl transferase (% of all reported adverse events)

Trebananib [50]

161
         

Arm 1

53

PTX + Trebananib 10 mg days 1, 8, 15

37

7.2

22.5

[n = 52] 12% hypokalemia, 10% peripheral neuropathy, 6% VTE.

Arm 2

53

PTX + Trebananib 3 mg days 1, 8, 15

19

5.7

20.4

11% hypokalemia, 9% dyspnea, 4% VTE

Arm 3

55

PTX + placebo days 1, 8, 15

27

4.6

20.9

4% hypokalemia, 9% VTE

PARP inhibitors

Maintenance

Olaparib [58, 59]

265
         

Arm 1

129

Pt CT → observation


4.8

27.8

3.1% fatigue, 0.8% anemia

Arm 2

136

Pt CT → Olaparib


8.4 **

29.8

7.7% fatigue, 5.1% anemia

BRCA (+)

Arm 1

62

Pt CT → observation
 
4.3

31.9
 

Arm 2

74

Pt CT → Olaparib
 
11.2 ***

34.9
 

BRCA (−)

Arm 1

61

Pt CT → observation
 
5.5

26.2
 

Arm 2

57

Pt CT → Olaparib
 
7.4 *

24.5
 

Recurrent disease

Olaparib [57]

97
         

Arm 1

32

Olaparib: 200 mg twice per day

25

6.5

13.1

6% abdominal pain, 6% constipation, 6% anemia

Arm 2

32

Olaparib: 400 mg twice per day

31

8.8

13.0

13% anemia, 9% fatigue, 6% nausea

Arm 3

33

PLD

18

7.1

13.0

38% Palmar-plantar erythrodysesthesia syndrome, 9% fatigue, 9% rash

Olaparib [60]

90
         

Arm 1

81

CP

58

9.6

37.6

4% fatigue 35% neutropenia, 7% anemia, 8% thrombocytopenia

Arm 2

81

CP + Olaparib

64 *

12.2 *

33.8

[n = 75] 7% fatigue 43% neutropenia, 9% anemia, 8% thrombocytopenia

Olaparib [61]

90
         

Arm 1

46

Olaparib

48

9.0


11% fatigue

Arm 2

44

Olaparib + Cediranib

80 *

11.7 *


41% HT, 27% fatigue, 23% diarrhea

Veliparib [63]

70
         

Arm 1

36

Cyclophosphamide

19.4

2.3


8% lymphopenia

Arm 2

34

Cyclophosphamide + Veliparib

11.8

2.1


35% lymphopenia

HER2 inhibitors

Recurrent disease

Pertuzumab [76]

130
         

Arm 1

65

GEM + placebo

5

2.6

13.1

22% neutropenia, 8% thrombocytopenia, 2% diarrhea, 2% back pain

Arm 2

65

GEM + Pertuzumab

14

2.9

13.0

35% neutropenia, 14% thrombocytopenia, 11% diarrhea, 9% back pain

Low HER3

Arm 1

35

GEM + placebo
 
1.4

8.4
 

Arm 2

26

GEM + Pertuzumab
 
5.3 **

12.5
 

Recurrent disease

Pertuzumab [77]

149
         

Arm 1

75

CP or CG

59

40.0 weeks

NR

Adverse events during the first six cycles of treatment were similar in both arms

Arm 2

74

CP or CG + Pertuzumab

61

34.1 weeks

28.2
 

Folate receptor

Recurrent disease

Vintafolide [91]

149
         

Arm 1

49

PLD

12

2.7


[n = 50] 2% Palmar-plantar erythrodysesthesia syndrome, 6% fatigue, 2% abdominal pain, 4% stomatitis, 10% neutropenia, 8% anemia, 9% leukopenia

Arm 2

100

PLD + Vintafolide → Vintafolide

18

5.0 *


[n = 107] 11% Palmar-plantar erythrodysesthesia syndrome, 9% fatigue, 8% abdominal pain, 8% stomatitis, 23% neutropenia, 9% anemia

FT inhibitor

First-line treatment

Lonafarnib [92]

105
         

Arm 1

52

CP


17.8

47.3

4% diarrhea

Arm 2

53

CP + Lonafernib → Lonafernib


14.2

33.4

23% diarrhea

IV, >1.0 cm b
           

Arm 1

14

CP
 
16.4

43.4
 

Arm 2

18

CP + Lonafernib → Lonafernib
 
11.5 *

20.6 *
 

ETA-receptor antagonist

Recurrent disease

Zibotentan [93]

120
         

Arm 1

61 (58)

CP + placebo → placebo

59

10.0


[n = 58] 31% neutropenia, 9% anemia, 16% leukopenia, 9% thrombocytopenia

Arm 2

59 (55)

CP + Zibotentan → Zibotentan

38 *

7.6


[n = 58] 41% neutropenia, 12% anemia, 10% leukopenia, 5% thrombocytopenia

PKCβ inhibitor

First-line treatment

Enzastaurin [94]

142
         

Arm 1

73 (18)

CP + placebo → placebo

39

15.2

47.3

[n = 72] 1% hypersensitivity, 3% constipation, 3% fatigue

Arm 2

69 (14)

CP + Enzastaurin → Enzastaurin

43

18.9

33.4

[n = 67] 1% constipation, 1% diarrhea, 3% dyspnea, 3% edema

Methyltransferase inhibitor

Recurrent disease

Decitabine [95]

29
         

Arm 1

14

Carboplatin

64

6.9


15% neutropenia

Arm 2

4

Carboplatin + Decitabine: 90 mg/m2

20 d *

1.9


60% neutropenia d

Arm 3

11

Carboplatin + Decitabine: 45 mg/m2
 
6.0

 

Plk inhibitor

Recurrent disease

Volasertib [96]

109
         

Arm 1

55

Non-Pt CT c

13

8.4 weeks


5% neutropenia, 2% anemia, 4% thrombocytopenia

Arm 2

54

Volasertib

15

13.1 weeks


44% neutropenia, 15% anemia, 17% thrombocytopenia, 17% leukopenia


PFS progression-free survival, M months, OS overall survival, PARP poly (ADP-ribose) polymerase, HER2 human epidermal growth factor receptor, BRCA (+) mutated BRCA, BRCA (−) wild-type BRCA, FT farnesyltransferase, ET endothelin, PKC protein kinase C, Plk Polo-like kinase, PtTx CT platinum- and taxane-based chemotherapy, PTX paclitaxel, Pt CT platinum-based chemotherapy, PLD pegylated liposomal doxorubicin, CP carboplatin plus paclitaxel, GEM gemcitabine, CG carboplatin plus gemcitabine, NR not yet reached, GI gastrointestinal, HT hypertension, VTE venous thrombosis

aNumber of patients eligible (Number of patients evaluable for response)

bStage IV and residual disease >1.0 cm

cInvestigator selected single-agent, non-platinum chemotherapy (pegylated liposomal doxorubicin, gemcitabine, weekly paclitaxel, or topotecan)

dCombination of arm 2 and 3

*P < 0.05, ** P < 0.001, *** P < 0.0001 vs. control arm

A phase I–II study of aflibercept in combination with docetaxel was conducted in patients with recurrent ovarian cancer [30]. The objective response rate was 54%, and grade 1–2 hypertension (11%) and grade 2 hypotension (2%) were adverse events specifically associated with aflibercept. Therefore, the combination of aflibercept and docetaxel seems safe and active for patients with recurrent ovarian cancer.


3.2.1.3 Cediranib


Cediranib is a highly potent, small-molecule, oral tyrosine kinase inhibitor of all three VEGF receptors (VEGFR1–3) and c-Kit, which competes for the ATP-binding site within the receptor kinase domain [31, 32]. A phase II trial of cediranib in patients with recurrent ovarian cancer reported a partial response (PR) rate of 17% and median PFS of 5.4 months [33]. These promising results led to a phase III study (ICON6) of patients with platinum-sensitive recurrent disease (Table 3.1) in which the median PFS was significantly prolonged in the platinum-based chemotherapy plus concurrent and maintenance cediranib arm (arm 3) vs. the chemotherapy and placebo (arm 1) (11.0 vs. 8.7 months) [34]. Although the OS analysis is ongoing, early median OS durations for arms 1 and 3 were 21.0 months and 26.3 months, respectively (P = 0.11).


3.2.1.4 Pazopanib


Pazopanib is a potent, selective oral multi-targeted receptor tyrosine kinase inhibitor of VEGFR1–3, platelet-derived growth factor receptor (PDGFR)-α and PDGFR-β, and fibroblast growth factor receptor (FGFR) 1–3 [35]. A phase II study (VEG104450) of pazopanib in patients with recurrent ovarian cancer reported a PR rate of 18% [36]. In a phase III trial (AGO-OVAR16), patients with International Federation Gynecology Obstetrics (FIGO) stage II–IV ovarian cancer received maintenance pazopanib or placebo for up to 24 months (Table 3.1) [37]. PFS was significantly prolonged for patients in the maintenance pazopanib arm vs. those in the placebo arm (median PFS: 17.9 vs. 12.3 months), although OS did not differ significantly between the arms at the interim analysis. In a randomized phase II trial (MITO 11), patients with platinum-resistant ovarian cancer received weekly paclitaxel with or without pazopanib; PFS was significantly longer in the paclitaxel/pazopanib group vs. the paclitaxel-only group (median PFS: 6.35 vs. 3.49 months) (Table 3.2) [38]. Two randomized phase II trials to evaluate chemotherapy (paclitaxel or gemcitabine) and combined effects with pazopanib are ongoing in patients with platinum-resistant ovarian cancer.


3.2.1.5 Nintedanib


Nintedanib (BIBF 1120) is a potent, oral tyrosine kinase inhibitor of VEGFR1–3, PDGFR-α and -β, and FGFR1–3. In a placebo-controlled randomized phase II trial of post-chemotherapy maintenance therapy in patients with relapsed ovarian cancer, nintedanib was well tolerated and associated with a potential improvement in PFS (Table 3.2) [39]. A phase III trial (AGO-OVAR12) investigated the combination of standard chemotherapy (paclitaxel/carboplatin) with nintedanib or placebo in patients with newly diagnosed FIGO stage IIB–IV ovarian cancer (Table 3.1) [40] and observed a significantly longer median PFS in the nintedanib group vs. the placebo group (17.2 vs. 16.6 months). The efficacy of nintedanib was particularly notable in patients with a low postsurgical disease burden (FIGO stage IIB–III, ≤1 cm residual postoperative tumor). Although the OS results are pending, further studies are needed to assess the clinical value of nintedanib, particularly in cohorts with lower tumor burdens.


3.2.1.6 Sunitinib


Sunitinib is a potent, oral multi-tyrosine kinase inhibitor that targets VEGFR1–3, PDGFR-α and -β, Flt-3, and c-Kit [41]. Three phase II trials were conducted to evaluate the efficacy and safety of this inhibitor in patients with recurrent ovarian cancer (Table 3.2) [4244]. However, efficacy seemed to be limited, with response rates of 3–17%, and the common adverse events included hypertension, gastrointestinal events, fatigue, and hand–foot syndrome.


3.2.1.7 Sorafenib


Sorafenib is an oral bis-aryl urea that inhibits c-Raf and b-Raf kinases and VEGFR-2 and -3, PDGFR-β, Flt-3, and c-Kit [45]. In a phase II trial (GOG-0170F) of sorafenib for patients with recurrent ovarian cancer, PR rate of 3% and median PFS and OS of 2.1 months and 16.3 months, respectively, were achieved [46]. However, a randomized phase II study of sorafenib maintenance therapy observed no significant difference in PFS between the sorafenib and placebo arms (Table 3.2) [47].


3.2.1.8 Trebananib


In tumor angiogenesis, angiopoietin-1 and angiopoietin-2 interact with the tyrosine kinase with immunoglobulin-like and EGF-like domains (Tie) 2 receptor, which is expressed on endothelial cells, to mediate blood vessel maturation and stabilization in a VEGF axis-independent pathway [48]. Trebananib (AMG 386), a neutralizing peptibody (i.e., peptide-Fc fusion protein), blocks the binding of both angiopoietin-1 and angiopoietin-2 to the Tie2 receptor, thereby inhibiting angiogenesis [49].

In a randomized phase II trial, trebananib combined with weekly paclitaxel prolonged PFS in patients with recurrent ovarian cancer (Table 3.2) [50]. A phase III trial, Trebananib in Ovarian Cancer-1 (TRINOVA-1), investigated trebananib in addition to single-agent weekly paclitaxel for patients with recurrent ovarian cancer (Table 3.1) [51]. Median PFS was significantly longer in the paclitaxel/trebananib group vs. the paclitaxel/placebo group (7.2 vs. 5.4 months), although the median OS did not statistically differ. Two subsequent phase III trials are ongoing: TRINOVA-2, which evaluates trebananib plus PLD for recurrent, partially platinum-sensitive ovarian cancer, and TRINOVA-3, which investigates trebananib plus first-line chemotherapy (carboplatin/paclitaxel) for FIGO stage III–IV ovarian cancer.



3.2.2 Targeting DNA Repair Mechanisms: Poly(ADP-Ribose) Polymerase (PARP)


PARPs have multiple functions, including DNA repair, cell dysfunction and necrosis, and inflammation (Fig. 3.2) [52]. PARP-1, the most abundant nuclear isoform, plays a vital role in DNA single-strand break (SSB) repair through the base excision repair pathway, whereas residual PARP activity (approximately 10%) is attributed to PARP-2. PARP inhibition causes an accumulation of DNA SSBs and consequent DNA double-strand breaks (DSBs) at replication forks. In normal cells, such DSBs are generally repaired by the BRCA1- and BRCA2-dependent homologous recombination (HR) DNA repair pathway. However, these lesions are not repaired in BRCA1- or BRCA2-deficient tumor cells, leading to genomic instability and cell death despite the existence of an alternate non-homologous end-joining pathway for DSB repair.

A416246_1_En_3_Fig2_HTML.gif


Fig. 3.2
Effect of DNA repair systems on poly(ADP-ribose) polymerase activity. Single-strand breaks lead to the activation of poly(ADP-ribose) polymerases (PARPs). PARP plays a key role in the repair of single-strand breaks. Treatment with a PARP inhibitor induces double-strand breaks and selectively kills homologous recombination-deficient tumor cells. BRCA breast cancer susceptibility gene

Female carriers of germline mutations in BRCA1 on chromosome 17q21 or BRCA2 on chromosome 13q31 have a higher risk of breast and ovarian cancer development. The lifetime risks of ovarian cancer are 54% for BRCA1 and 23% for BRCA2 mutation carriers [53]. Although germline mutations in those genes are seen in 5–10% of all ovarian cancer patients, a loss of HR function (BRCAness), either via genetic or epigenetic events in BRCA1 or BRCA2 or alterations in other genes (e.g., EMSY, PTEN, RAD51C, ATM, ATR, Fanconi anemia genes), are observed in approximately half of high-grade serous ovarian carcinomas [54]. In ovarian cancer, a BRCAness profile may correlate with responses to platinum-based chemotherapy and PARP inhibitors.


3.2.2.1 Olaparib


Olaparib is an oral small-molecule PARP inhibitor that induces synthetic lethality in cells with defective BRCA function [55]. Pooled data from phase I/II trials of olaparib (400 mg twice daily) monotherapy demonstrated an objective response of 36% in germline BRCA1/2 mutation carriers with recurrent ovarian cancer [56, 57]. An ongoing phase III trial of BRCA mutation carriers with platinum-sensitive recurrent ovarian cancer, SOLO3, compares olaparib monotherapy vs. the physician’s selected chemotherapy (weekly paclitaxel, topotecan, PLD, or gemcitabine).

The efficacy of olaparib maintenance therapy was evaluated in a randomized phase II study of patients with platinum-sensitive, relapsed, high-grade serous ovarian cancer (Table 3.2) [58, 59]. Among BRCA mutation carriers, the median PFS was significantly longer in the olaparib group vs. the placebo group (11.2 vs. 4.3 months), and similar results were observed for wild-type BRCA carriers (7.4 vs. 5.5 months). However, OS did not significantly differ between the groups. Phase III confirmatory trials of maintenance olaparib monotherapy are ongoing in BRCA mutation carriers with ovarian cancer after first-line platinum-based chemotherapy (SOLO1) and those who have achieved a complete response (CR) or PR with platinum chemotherapy (SOLO2).

A combination therapy of olaparib with chemotherapy (carboplatin/paclitaxel) was tested in patients with platinum-sensitive recurrent, high-grade serous ovarian cancer in a randomized phase II trial (Table 3.2) [60]. PFS was significantly longer in patients treated with olaparib plus chemotherapy followed by maintenance olaparib monotherapy vs. those treated with chemotherapy alone (median PFS, 12.2 vs. 9.6 months), although OS did not differ significantly between the treatment groups. The combined effect of olaparib with targeted agents on patient outcome is also under investigation. In a randomized phase II trial, recurrent platinum-sensitive ovarian cancer patients received olaparib alone or cediranib plus olaparib (Table 3.2) [61]. The median PFS was significantly improved in the combination group vs. the olaparib alone group (17.7 vs. 9.0 months). However, grade 3/4 adverse events were more common with combination therapy. These promising results initiated a randomized phase III trial (NRG-GY004) of platinum-sensitive recurrent ovarian cancer with three treatment arms: (1) carboplatin and paclitaxel (regimen I), gemcitabine (regimen II), or PLD (regimen III), (2) olaparib, and (3) olaparib and cediranib.


3.2.2.2 Other PARP Inhibitors


Besides olaparib, several PARP inhibitors, including veliparib, rucaparib, and niraparib, are being evaluated in clinical trials. Veliparib was tested as a monotherapy for BRCA-mutated recurrent ovarian cancer in a phase II trial (GOG-0280), yielding an overall response rate of 26% [62]. A randomized phase II trial of veliparib with low-dose cyclophosphamide did not improve the response rate or median PFS in patients with high-grade serous ovarian cancer (Table 3.2) [63]. A phase III trial of veliparib with first-line chemotherapy (carboplatin/paclitaxel) followed by maintenance veliparib (GOG-3005) is currently recruiting patients with high-grade serous ovarian cancer. Rucaparib (ARIEL3 trial) and niraparib (NOVA trial) are also currently under evaluation in phase III trials of maintenance treatment after platinum-sensitive recurrent ovarian cancer. These trials are recruiting both sporadic and BRCA-mutated ovarian cancer patients.


3.2.3 Targeting the Human Epidermal Growth Factor Receptor Family


The epidermal growth factor receptor (EGFR; HER in humans) family comprises four distinct transmembrane tyrosine kinase receptors: HER-1 (EGFR/erbB1), HER-2/neu (erbB2), HER-3 (erbB3), and HER-4 (erbB4) [64]. These receptors are activated via C-terminal autophosphorylation by ligand binding (although HER2 has no known ligand) and multiple receptor homo- or hetero-dimerization combinations, thus triggering downstream signaling pathways such as the MAPK and PI3K/Akt pathways and thus inducing cancer-cell proliferation, blocking apoptosis, activating invasion and metastasis, and stimulating tumor-induced neovascularization. Accordingly, HERs are attractive targets for anticancer therapies (Fig. 3.3) [64].

A416246_1_En_3_Fig3_HTML.jpg


Fig. 3.3
Targeting the human epidermal growth factor receptor family members and their downstream signaling pathways in gynecologic cancer. The human epidermal growth factor receptor (HER) family consists of four distinct transmembrane tyrosine kinase receptors, and receptor-specific ligands selectively bind to each of them. The receptor undergoes homo- or hetero-dimerization that leads to receptor autophosphorylation that activates a series of downstream signaling pathways, such as mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt pathways that control cell growth and apoptotic signaling. EGFR epidermal growth factor receptor, HER human EGFR, PTEN phosphatase and tensin homolog, mTOR mammalian target of rapamycin, S6K1 ribosomal protein S6 kinase 1, 4E-BP1 eukaryotic translation initiation factor 4E binding protein 1, P phosphate

HER family members are expressed in many human malignancies, including ovarian cancers, in which a wide range of HER family expression has been reported [EFGR, 4–100% (average, 48%); HER-2, 0–100% (average, 40%); HER-3, 3–90% (average, 48%); and HER-4, 45–92% (average, 71%)] [65]. Overexpression of HER, particularly EGFR and HER-2, may correlate with poor prognosis and decreased therapeutic response, although clinical data are contradictory. Several EGFR and HER-2 inhibitors have been tested in patients with ovarian cancer.


3.2.3.1 EGFR Inhibitors


The EGFR tyrosine kinase inhibitors erlotinib and gefitinib have been tested in phase II trials, which observed limited activities of these agents as monotherapies for recurrent ovarian cancer (response rates, 6% and 0–4%, respectively) [6668]. The European Organization for Research and Treatment of Cancer-Gynecological Cancer Group (EORTC-GCG) conducted a phase III study of erlotinib (EORTC 55041) in patients with ovarian cancer after first-line, platinum-based chemotherapy (Table 3.1) [69]. Unfortunately, maintenance erlotinib for 2 years after first-line treatment did not improve PFS or OS in these patients.

The monoclonal EGFR-specific antibodies cetuximab and matuzumab block the binding of EGF to its receptor, thus inhibiting ligand-induced receptor autophosphorylation. Both cetuximab and matuzumab were tested in patients with recurrent ovarian cancer in phase II settings, with overall response rates of 4% and 0%, respectively [70, 71]. A phase II trial (GOG-0146P) assessed cetuximab activity in combination with carboplatin for EGFR-positive, recurrent platinum-sensitive ovarian cancer but reported only modest activity, with a PR rate of 35% [72]. Similarly, a phase II trial of cetuximab with carboplatin/paclitaxel as a first-line treatment for FIGO stage III/IV ovarian cancer did not demonstrate PFS prolongation when compared with historical data [73].


3.2.3.2 HER2 Inhibitors


Humanized monoclonal HER2 antibodies, trastuzumab and pertuzumab, were evaluated in phase II trials, which reported limited activity of these agents as monotherapies for recurrent ovarian cancer [74, 75]. A combination of pertuzumab with gemcitabine was tested in a phase II trial of platinum-resistant ovarian cancer patients (Table 3.2) [76] who were randomly allocated to gemcitabine plus placebo or pertuzumab, with objective response rates of 5% and 14%, respectively. Among patients whose tumors exhibited low HER3 mRNA expression, the median PFS was significantly longer with pertuzumab vs. placebo (5.3 vs. 1.4 months), although increased grade ≥ 3 neutropenia, diarrhea, and back pain were observed in the former. Pertuzumab was also evaluated together with carboplatin-based chemotherapy in a randomized phase II study of patients with platinum-sensitive, recurrent ovarian cancer (Table 3.2) [77]. No significant differences in PFS or OS were observed between chemotherapy (carboplatin and either paclitaxel or gemcitabine) alone and chemotherapy with pertuzumab. Unfortunately, no differences were observed between the arms in a biomarker analysis of HER3 mRNA expression. These studies suggest that pertuzumab, in combination with chemotherapy, is mainly effective in patients with platinum-resistant ovarian cancer and low HER3 mRNA expression.


3.2.3.3 Other HER Family Inhibitors


Phase II trials of single-agent targeted therapies, including the HER family tyrosine kinase inhibitors lapatinib and canertinib, have shown only modest efficacy [78, 79]. Lapatinib, a dual tyrosine kinase inhibitor of EGFR and HER2, was evaluated in recurrent ovarian cancer patients, although no objective responses were observed [78]. The combination therapy of lapatinib plus topotecan was also tested in a phase II trial in patients with platinum-resistant ovarian cancer, but lacked sufficient activity (no CR, one PR) [80]. Another phase II trial evaluated a pan-HER family tyrosine kinase inhibitor, canertinib, in patients with platinum-resistant recurrent ovarian cancer [79]; although two oral doses of canertinib (50 mg and 200 mg) were evaluated, no responses were observed.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Oct 17, 2017 | Posted by in GYNECOLOGY | Comments Off on Signal Transduction and Targeted Therapy for Gynecologic Cancer

Full access? Get Clinical Tree

Get Clinical Tree app for offline access