Skip to main content
Molecular Therapy Oncolytics logoLink to Molecular Therapy Oncolytics
. 2023 Feb 19;28:293–306. doi: 10.1016/j.omto.2023.02.006

Targeting receptor tyrosine kinases in ovarian cancer: Genomic dysregulation, clinical evaluation of inhibitors, and potential for combinatorial therapies

Ying Wei 1,9, Sonia Erfani 2,3,9, David Schweer 4,9, Rafael de Gouvea 2,5,9, Javeria Qadir 6,9, Junfeng Shi 2,4,7,9, Kai Cheng 8, Dabao Wu 1, Rolf Craven 2, Yadi Wu 2,4, Thibault Olivier 2, Lauren A Baldwin 4, Binhua Zhou 4, Ying Zhou 1,, Weidong Zhao 1,∗∗, Burton B Yang 6,∗∗∗, Frederick R Ueland 4,∗∗∗∗, Xiuwei H Yang 2,∗∗∗∗∗
PMCID: PMC9999170  PMID: 36911068

Abstract

Epithelial ovarian cancer (EOC) remains one of the leading causes of cancer-related deaths among women worldwide. Receptor tyrosine kinases (RTKs) have long been sought as therapeutic targets for EOC, as they are frequently hyperactivated in primary tumors and drive disease relapse, progression, and metastasis. More recently, these oncogenic drivers have been implicated in EOC response to poly(ADP-ribose) polymerase (PARP) inhibitors and epigenome-interfering agents. This evidence revives RTKs as promising targets for therapeutic intervention of EOC. This review summarizes recent studies on the role of RTKs in EOC malignancy and the use of their inhibitors for clinical treatment. Our focus is on the ERBB family, c-Met, and VEGFR, as they are linked to drug resistance and targetable using commercially available drugs. The importance of these RTKs and their inhibitors is highlighted by their impact on signal transduction and intratumoral heterogeneity in EOC and successful use as maintenance therapy in the clinic through suppression of the VEGF/VEGFR axis. Finally, the therapeutic potential of RTK inhibitors is discussed in the context of combinatorial targeting via co-inhibiting proliferative and anti-apoptotic pathways, epigenomic/transcriptional programs, and harnessing the efficacy of PARP inhibitors and programmed cell death 1/ligand 1 immune checkpoint therapies.

Keywords: ovarian cancer, receptor tyrosine kinases, EGFR, ERBB2, MET, VEGFR, targeted therapy

Graphical abstract

graphic file with name fx1.jpg


In this review, Wei et al. summarize the current landscape of the receptor tyrosine kinases (RTKs) with regard to their biological role in epithelial ovarian cancer (EOC) and druggable targets, highlighting the potential of combinatorial targeted therapy as a mean of overcoming resistance to the contemporary therapies.

Introduction

Epithelial ovarian cancer (EOC) accounts for more than 90% of human ovarian cancer cases and is one of the leading causes of cancer-related deaths among women worldwide.1,2 In 2021, there were approximately 13,000 EOC-related deaths in the United States alone.3 Alarmingly, the majority (70%) of these patients initially present with advanced disease and face 5-year survival rates as low as 50%. The mainstay of management of EOC involves surgical debulking combined with systemic platinum-/taxane-based chemotherapy in either a neoadjuvant or adjuvant setting.4,5,6 Despite diverse targeted therapies aiming to cure EOC, 75% of women still experience a relapse or recurrence of the disease.7,8 Hence, more effective therapies and treatment regimens are needed to improve patient survival and outcomes.

Conventional platinum- and taxane-based chemotherapies are known to disrupt EOC via impairing microtubule-mediated cell division and DNA synthesis/replication process, respectively. Poor patient response to such treatment regimens is increasingly linked to broad heterogeneity among EOC tumors in terms of oncogenic activation or dysregulation at cellular, genetic, and epigenetic levels.3,9 To date, 75% of the EOC diagnosed in the clinic belong to the high-grade serous ovarian cancer (HGSOC) subtype. HGSOC is further stratified into four major subgroups: proliferative, mesenchymal, immunoreactive, and differentiated, which markedly differ in their cellular origin, morphology, and composition, as well as response to the current therapies.1 There are also less common histological subtypes of EOC, including low-grade serous ovarian cancer, mucinous, clear cell, and endometroid.1,9 Because of high intrinsic genomic instability, EOC tumors possess unusually high numbers of oncogenic mutations, as well as extensive rearrangements of chromosomal segments, causing gene deletion, amplification, and fusion.9,10,11,12,13,14 In addition, a high degree of intratumoral heterogeneity in EOC tumors occurs at genetic and epigenetic levels.15,16 These lines of dysregulation empower ovarian tumors to counteract traditional chemotherapies, and to fuel disease recurrence and shorter duration of patient survival.10,17,18,19 Importantly, they also expose various vulnerabilities to diverse therapeutic targeting.

A variety of molecular targeting strategies have been pursued for EOC treatment, ranging from direct inhibition of the oncogenic pathways to co-targeting of molecular machinery involved in chromatin remodeling and repair of damaged DNA.1,19,20,21,22,23 Of high promise to clinical application, however, is the disruption of oncogenic signaling of receptor tyrosine kinases (RTKs) and their downstream effectors, which are extensively dysregulated in nearly half of the HGSOC tumors, according to large-scale genomic analyses of patient biopsies.11,19 Through elevated transcription or protein translation, RTKs become highly expressed on the surface of tumor cells. Upon ligand-based stimulation or overexpression they undergo clustering/aggregation and autophosphorylation to become activated and to allow the recruitment of diverse substrates and assembly of the multi-functional protein complexes. These changes, in turn, stimulate the pro-proliferative and/or anti-apoptotic pathways and expression of cell mitosis-related genes.24 Meanwhile, they also lead to reprogramming of cell-cell and cell-extracellular matrix (ECM) adhesion toward epithelial-mesenchymal transition (EMT) phenotype, avoidance of programmed cell death, and immune surveillance.24 Conceivably, ovarian carcinomas driven by various RTK oncogenes are targetable via their small-molecule inhibitors or function-blocking antibodies.25

To date, a number of RTK antagonists have been explored as potential monotherapy or part of the combination therapies for EOC.26,27,28,29,30 Particularly, they have been evaluated through pairing with other treatment options, notably poly(ADP-ribose) polymerase (PARP) inhibitors used to treat a subgroup of EOC patients carrying genetically inherited mutations or low expression of the BRCA1/2 gene.31,32,33 Extensive studies have found that ovary and other tissues/organs from these patients are highly dependent on the PARP-driven pathway for repairing damaged DNA, avoidance of senescence and apoptosis or death.26,34 These findings prompt the development of contemporary PARP inhibitor-based therapy for such a group of EOC patients.35,36,37 Intriguingly, when prolonged PARP inhibitor treatment is applied, some RTKs seem to become activated in a feedback manner.38 Moreover, RTK targeting is viewed valuable to EOC treatment from the angle of harnessing the efficacy of immune checkpoint based or inhibitors of the epigenetic regulators such as azacytidine or bromodomain and extra-terminal (BET) inhibitors.39,40,41,42

With accumulating evidence on the impact of the ERBB family, a subgroup of druggable RTKs, on disease progression and drug resistance, this review concentrates on therapeutic evaluation of these RTKs and their antagonists for EOC. Given the growing promise of combinatorial therapy for EOC, we have discussed the scope and efficacy of ERBB inhibitors in the wake of co-targeting with various chemo- and targeted therapies such as PARP inhibitors and programmed cell death 1/ligand 1-based immune checkpoint therapies.

Oncogenic activation and signal transduction of the ERBBs

A glance at protein structure, enzymatic activity, and oncogenic activation

While many RTKs exist in the human kinome, members of the ERBB family are the most frequently dysregulated oncogenic drivers across human epithelial cancers, including EOC.24,43,44,45 As the prototype of druggable RTKs, the ERBB family consists of four members: EGFR (ERBB1/HER1), ERBB2/HER2, ERBB3/HER3, and ERBB4/HER4.24 Structurally, members of the ERBB family are composed of an extracellular ligand-binding domain, a cell membrane-spanning region, and an intracellular tyrosine kinase domain (Figure 1). Within the family, only EGFR and ERBB4 become activated through their extracellular domain interaction with the ligands, such as epidermal growth factor, transforming growth factor α, amphiregulin, and heregulin.46 All the family members, with the exception of ERBB3, possess enzymatic activities and are activated upon homodimerization/polymerization or heterodimerization with ERBB3 or the ligand-activated form of EGFR, ERBB4, or c-Met on the cell surface.24 Thus, in human carcinomas, EGFR, ERBB2, and ERBB4 frequently become oncogenic once amplified, mutated, or overexpressed at the genomic or protein level, but rarely through structural alteration or gene fusion.47

Figure 1.

Figure 1

Schematic illustration of the signaling and functional roles of the major druggable RTKs in epithelial ovarian tumor cells

Physiological ligands and dimerization (homo- and hetero-), signaling pathways and cellular roles are highlighted for ERBB receptors and other RTKs. Ligands for some common RTKs are listed in the top left corner and include the following: EGF, epidermal growth factor; HB-EGF, heparin-binding EGF-like growth factor; HGF, hepatocyte growth factor; PARP, poly(ADP-ribose) polymerase inhibitors; TGF-α, transforming growth factor α.

Like most of RTKs, the pro-tumorigenic and pro-metastatic roles of ERBB receptors are achieved by eliciting arrays of downstream signaling cascades (Figure 1), highlighted by the activation of PI3K/Akt/mTOR and Ras/Raf/MEK/ERK pathways, PLC-γ1, STATs, and Src.24 Aside from promotion of nutrient uptake and biosynthesis (e.g., glucose, amino acids, and nucleotides), these signaling cascades markedly upregulate expression of many pro-proliferative and pro-cell survival genes at the epigenetic, transcriptional, and post-transcriptional levels.48,49,50 It is also worth noting that members of the ERBB family and c-Met impact tumor growth and progression via intricate crosstalk with other mediators such as inflammation- and integrin-dependent signaling or epigenetic machinery.51,52,53,54 Inhibiting their signaling cascades leads to cell-cycle arrest, apoptosis, impaired tumor cell growth, and pro-invasive behavior.30,51 ERBB receptors are also known as crucial players in tumor metastasis, as they promote EMT, tumor invasiveness, angiogenesis, and distant metastatic progression. 55

There is evidence that RTKs become activated through a feedback loop in tumor cells after prolonged treatment with traditional chemotherapies or targeted therapies. Notably, several activated RTKs were detected upon sustained inhibition of the RAS/RAF/MEK pathway or transcription/epigenetic mediator BRD4, a member of the BET family.40,56 In line with this evidence, FAK, an integrin-linked non-RTK and a key downstream signaling effector of ERBB2 and other RTKs, modulates tumor cell responses to pharmacological inhibition of BRD4, according to our recent study.23,52,57 In addition, RTKs may contribute to poor prognosis and drug resistance by promoting expression or activity of the drug transporters.58,59 In these scenarios, RTK antagonists could serve as a second line of therapy for EOC patients after acquiring drug resistance.

Genomic dysregulation and signaling alteration of the druggable ERBBs in EOC tumors

Members of the ERBB family have been strongly implicated as key drivers of EOC malignancy (Figure 1).24 Notably, EGFR is overexpressed in 30%–98% of EOC and is linked to poor clinical outcomes.60,61 ERBB2 is overexpressed or amplified in about 20%–66% of the biopsies across multiple EOC patient cohorts.62,63 The dysregulation of ERBB receptors and other RTKs in EOC is also readily detectable at the mRNA level (Figure 2), in contrast to the point mutations in their cytoplasmic domains of RNA splicing as seen in other cancer types (e.g., EGFR in glioblastoma).51 Interestingly, the genomic and mRNA alterations seem mutually exclusive for c-MET and the ERBB receptors (Figure 2). Importantly, the altered gene copy number or expression of the ERBB family possesses diagnostic value. In particular, ERBB2 expression correlates with poor prognosis of EOC.64 In addition, despite the lack of independent kinase activity, the ERBB3 gene is frequently amplified in EOC, and this alteration correlates with poor progression-free survival (PFS).43,65,66 Exceptionally, ERBB4 forms a fusion gene with IKZF2 in some EOC tumors.26

Figure 2.

Figure 2

Profiling analysis of genomic and mRNA alterations of the ERBB receptors and other druggable receptor tyrosine kinases in patients with ovarian serous cystadenocarcinoma (TCGA, Pan Cancer Atlas; patients/samples: n = 585)

EGFR, ERBB2, and 26 other genes were profiled in terms of fusions, mutations, protein expression (Z scores [RPPA]), putative copy-number alterations from GISTIC, mRNA expression (relative to all samples, log RNA Seq V2 RSEM).

Besides genomic alterations, the importance of the ERBB receptors in EOC is underscored by frequent activation of their downstream signaling intermediates (Figure 3). Notably, the PI3K/Akt/mTOR and Ras/Raf/MEK/ERK pathways, which are two common sets of pathways downstream of ERBB receptors, are constitutively activated in approximately 70% of ovarian tumors at rates second only to DNA repair pathways.19 This dysregulated signaling is oncogenic and drives tumor cell proliferation, migration, survival, invasion, and chemotherapy resistance.67,68 In addition, ERBB receptors are implicated in regulating DNA damage response and disease progression in EOC.64 Collectively, in EOC tumors, members of the ERBB family undergo extensive genomic alterations or mutations, and their signaling pathways are markedly rewired or dysregulated, fueling their therapeutic utility.

Figure 3.

Figure 3

Profiling analysis of genomic and mRNA alterations of the druggable downstream effectors of the RTK-dependent signaling pathways in ovarian serous cystadenocarcinoma

The output was obtained through analysis of the TCGA cohort (Pan Cancer Atlas, patients/samples: n = 585) described in Figure 2.

Other RTKs in EOC

Besides ERBB receptors, other RTKs, including c-Met, AXL, PDGFR, members of the EPH family, and VEGFR, are regarded as potential valuable targets for EOC treatment (Figure 2). These RTKs are activated in mechanisms similar to the ERBB receptors, presumably due to their structural similarity.33,56,69,70 In particular, c-Met activation is implicated as part of a feedback loop under prolonged exposure to traditional chemotherapies or targeted therapies. In addition, there is a feedback-type activation of several druggable RTKs implicated during sustained inhibition of the RAS/RAF/MEK pathway or transcription/epigenetic modulators such as BRD4.40,56

Clinical evaluation of the ERBB antagonists as monotherapy

Thus far, a number of ERBB antagonists, including humanized monoclonal antibodies and small-molecule kinase inhibitors, have been tested as monotherapy for EOC (Table 1). Mechanistically, these agents act via two distinct modes: (1) interference of the extracellular ligand binding or receptor dimerization and (2) competitive inhibition of the ATP-binding capability of the kinase domain (Figure 1). In the first mode, monoclonal antibody drugs block the ligand binding of the ERBB receptors on the cell surface or facilitate receptor internalization and degradation to prevent clustering/aggregation-dependent oncogenic activation. In the second mode, small-molecule-based tyrosine kinase inhibitors (TKIs) directly impair autophosphorylation and kinase activities of the ERBB receptors. Upon treatment with these inhibitors, signal transduction of the ERBB oncogenes is blocked, leading to diminished protein translation and transcription for many critical genes involved in nutrient supply, cell proliferation, and survival.60 Given the marked difference in their modes of action, the function-blocking antibodies and TKIs may exert divergent anti-tumor effects, making them attractive candidates for combinatorial targeting therapies.36

Table 1.

A selected list of the clinical trials with ERBB-targeted agents as monotherapy in ovarian/genitourinary cancers

Target Intervention Clinical setting Trial ID Phase Enrollment
EGFR gefitinib recurrent or persistent ovarian epithelial cancer or primary peritoneal cancer NCT00023699 II 30
erlotinib epithelial ovarian, primary peritoneal or fallopian tube cancer (high-risk stage I or stage II–IV) with responding or stable disease after first-line platinum-based chemotherapy NCT00263822 III 835
erlotinib recurrent metastatic or unresectable non-small lung cancer, ovarian cancer, or squamous cell carcinoma of the head and neck NCT00063895 I/II 80
erlotinib persistent or recurrent squamous cell carcinoma of the cervix NCT00031993 II 51
matuzumab recurrent ovarian cancer following treatment for primary or secondary platinum-refractory disease with evidence of tumor EGFR (HER1) expression NCT00073541 II 38
ERBB2 lapatinib persistent or recurrent ovarian epithelial or peritoneal cancer NCT00113373 II 28
recurrent or persistent endometrial cancer NCT00096447 II 31
trastuzumab recurrent or persistent endometrial cancer NCT00006089 II 34

To date, many small-molecule inhibitors of ERBB2, EGFR, and MET have been clinically investigated for EOC, as these RTKs are frequently dysregulated in primary tumors according to analyses of the TCGA data (Figures 2 and 3) and other published patient cohorts.61 Notably, erlotinib, a small-molecule inhibitor of EGFR, has been shown to inhibit growth of EOC cells and to modulate their sensitivity to cytotoxic agents such as carboplatin.71 Lapatinib and poziotinib, two common inhibitors of ERBB2, have also been studied regarding their role in modulating EOC resistance to taxane-based regimens.72,73 In a more recent preclinical study, afatinib, a third-generation small-molecule inhibitor of ERBB oncogenes, was found to have moderate efficacy against EOC.74 Interestingly, these RTK inhibitors seem particularly effective against a subgroup of EOC cells overexpressing ERBB2 receptor.75 Based on this evidence, clinical trials with several inhibitors of EGFR and ERBB2 have been carried out in EOC patients (Table 1). Again, afatinib was highly effective against patients carrying ERBB2 mutations.76,77,78 Unexpectedly, lapatinib exhibited poor efficacy when used as a monotherapy in persistent or recurrent EOC.79

Besides small-molecule inhibitors, function-blocking monoclonal antibodies of EGFR and ERBB2 have been clinically tested in EOC (Table 1). These agents, including trastuzumab, pertuzumab, and matuzumab, inhibit activity of the ERBB receptors by interfering with their homo- or hetero-dimerization-mediated activation in tumor cells.24,43 In line with the notion, ovarian tumor cells expressing high levels of ERBB2 proteins are sensitive to the effect of trastuzumab, the first generation of humanized ERBB2 monoclonal antibody, and to taxane-based agents.80,81 Mechanistically, trastuzumab appears to exert a pro-apoptotic effect through inhibition of the PI3K/AKT axis.82 Also, pertuzumab, a humanized monoclonal antibody that disrupts the heterodimerization between ERBB2 and other members of the ERBB family, is shown to suppress growth of ovarian cancer cells both in vitro and in vivo.83,84 Furthermore, cetuximab, a monoclonal antibody against EGFR, was investigated as monotherapy for patients with primary peritoneal cancers or recurrent EOC.85 Unfortunately, this agent exhibited poor efficacy among patients with recurrent EOC.

Overall, the initial findings from the above clinical studies suggest that ERBB inhibitors may provide limited efficacy in EOC. Nevertheless, these agents could have potential value for EOC treatment if suitable biomarkers are developed to stratify the EOC patient populations according to expression level or activity of ERBB receptors.

Biomarkers and clinical evaluation of the ERBB inhibitors as part of combinatorial therapies

Aside from being evaluated as monotherapy, a number of ERBB inhibitors have been clinically pursued for EOC treatment in conjunction with conventional chemotherapies or other targeting agents. Notably, multiple clinical trials have been performed to evaluate efficacy of TKIs of multiple ERBB members for EOC (Table 1, Table 2 and 2). Interestingly, one of these trials is the phase II study (NCT00436644) investigating the impact of the combination of lapatinib and topotecan in patient populations with platinum-refractory/-resistant peritoneal or ovarian carcinomas (Table 2). Unfortunately, the data from analysis of correlative markers did not show dramatic predicted benefit. In addition, disruption of the ERBB signaling with lapatinib appears insufficient to overcome topotecan resistance.

Table 2.

A selected list of the clinical trials with combinations of ERBB-targeted agents and chemotherapy in ovarian/genitourinary cancers

Target Intervention Clinical setting Trial ID Phase Enrollment
EGFR gefitinib + tamoxifen epithelial ovarian cancer, cancer of the fallopian tube or peritoneum refractory or resistant to platinum- and taxane-based chemotherapy NCT00189358 (AGO-OVAR 2.6) II 49
topotecan then erlotinib topotecan-treated epithelial and/or serous ovarian cancer NCT01003938 II 6
erlotinib with docetaxel/carboplatin followed by maintenance therapy of erlotinib newly diagnosed stage III or IV epithelial ovarian, primary peritoneal cavity or fallopian tube cancer NCT00217529 I/II 30
carboplatin + erlotinib recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer for which no standard curative therapy exists NCT00030446 II 50
paclitaxel/carboplatin + erlotinib first-line treatment of stage III or IV ovarian, fallopian tube, or primary peritoneal cancer NCT00059787 II 56
cetuximab + carboplatin recurrent, platinum-sensitive epithelial ovarian or primary peritoneal cancer NCT00086892 II 29
ERBB2 lapatinib + topotecan platinum-refractory/resistant epithelial ovarian or primary peritoneal cancer NCT00436644 II 18
paclitaxel + pertuzumab + topotecan
paclitaxel + pertuzumab + placebo
gemcitabine + paclitaxel + pertuzumab + topotecan
gemcitabine + paclitaxel + placebo + topotecan
platinum-refractory/resistant epithelial ovarian, primary peritoneal, and/or fallopian tube cancer with low HER3 expression NCT01684878 (PENELOPE) III 208
placebo comparator: placebo + gemcitabine
active comparator: pertuzumab + gemcitabine
platinum-refractory/resistant ovarian, primary peritoneal, or fallopian tube carcinoma NCT00096993 II 131
experimental: chemotherapy (paclitaxel + gemcitabine + carboplatin) + pertuzumab
active comparator: chemotherapy (paclitaxel + gemcitabine + carboplatin)
platinum-sensitive recurrent ovarian cancer NCT02004093 II 149
ERBB3 experimental: seribantumab + paclitaxel
active comparator: paclitaxel alone
platinum resistant/refractory advanced ovarian cancer NCT01447706 II 233

In conjunction with systemic chemotherapeutic agents

The most clinically relevant trials of the ERBB antagonists are investigations of both drug efficacy and biomarkers for erlotinib-based combinatorial therapies. One such trial (NCT0030446) is a phase II study of the combination of erlotinib plus carboplatin in patients with recurrent EOC (Table 2).86 This drug combination appears effective for EOC patients with platinum-sensitive disease only. In a phase III trial (NCT00263822), an effort was made toward development of better biomarkers for analysis of erlotinib efficacy in a cohort of ovarian cancer patients who underwent first-line platinum-based treatment (Table 2).87 The results from this trial showed that the PFS and overall survival (OS) of the EOC patients were strongly associated with copy number gain of EGFR gene, as the patients carrying amplified EGFR exhibited poorer OS than their counterparts.87,88 This finding implies that delineating EGFR mutations alone may not be sufficient to foresee patient response to erlotinib treatment, nor do the gain-of-function mutations in EGFR-associated signaling cascades (e.g., KRAS, BRAF, NRAS, and PIK3CA). Furthermore, patient populations with at least one mutation in either KRAS, NRAS, BRAF, or PIK3CA exhibited longer PFS than those without any of these genetic mutations. Together, these trial results implicate an uphill challenge for pursuing clinical application of small-molecule inhibitors of ERBB receptors for EOC treatment, regardless of being used as monotherapy or in association with chemotherapies.89

Aside from small-molecule inhibitors, several RTK-blocking antibodies have been sought for EOC treatment in the context of combining with chemotherapies. Notably, in the PENELOPE trial (NCT01684878), adding pertuzumab did not significantly improve PFS in the patient cohort with platinum-resistant tumors and low expression of ERBB3.43 The results from a subgroup analysis, however, revealed better efficacy of pertuzumab treatment in conjunction with gemcitabine and paclitaxel.90 In addition, cetuximab was tested in combination with carboplatin for patients with recurrent ovarian cancer.85 It was also tested in combination with paclitaxel/carboplatin for the patient population with advanced-stage peritoneal lesions or fallopian tube cancer.91 Collectively, these trials indicate that cetuximab has some beneficial effect for patients with EGFR-positive and platinum-sensitive ovarian carcinomas.

Combination of chemical inhibitors and antibody blockers

The simultaneous administration of two inhibitors against the same ERBB target has also been explored for EOC treatment. This type of strategy is sometimes referred to as “dual blockade” as it disrupts oncogenic activity through simultaneous targeting of extracellular and intracellular domains of ERBB receptors. Notably, in one experimental study, the combination of afatinib and trastuzumab yielded an additive/synergistic effect on the ERBB2+ breast cancer cells.92 Intriguingly, this effect appeared independent of tumor cell sensitivity to trastuzumab. Instead, afatinib appears to act on tumor cell resistance to trastuzumab by blocking the compensatory signaling pathway. Whether this phenomenon also occurs in vivo or in the clinical setting, however, remains elusive. One major concern is the high prevalence of constitutive activation of the PI3K/Akt pathway in EOC tumors due to PIK3CA mutation or PTEN loss or Akt amplification, as highlighted in the TCGA patient population (Figure 3). This is also consistent with a recent genomic study.19 To some extent, knowing such genomic landscape may aid in foreseeing intrinsic resistance to the ERBB antagonist-based therapies in EOC patient population. It may also fuel the co-administration of PI3K or AKT inhibitors and trastuzumab as a new line of targeted therapy for EOC patients.30,93

Toward synthetic lethal targeting

In combination with PARP inhibitors

One of the most exciting advances in targeted therapy for EOC over the past decade is the clinical application of small-molecule inhibitors of PARP enzymes, which appears particularly effective in treatment of the patient population carrying functionally defective BRCA1/2 genes.3,8 Thus far, several RTK inhibitors have been explored for treatment of metastatic EOC in conjunction with the PARP inhibitors. Neratinib, which irreversibly inhibits activities of multiple members of the ERBB family through disruption of autophosphorylation and signal transduction, has been approved for use in treatment of metastatic breast cancer.94 This pan-ERBB inhibitor also seems capable of downregulating expression of ERBB1/2/4, c-MET, PDGFR, and mutant RAS proteins via the autophagic degradation route.95 In a recent study, the combination of neratinib with niraparib, a PARP inhibitor, was found to have a synergistic effect in cisplatin-resistant or multi-drug-resistant ovarian cancer cells.96 Mechanistically, this drug combination appears to induce the ATM-dependent activation of AMPK in tumor cells, which in turn alters signal transduction, autophagy induction and pro-tumorigenic roles of mTOR, ULK1, and ATG13.97 There is an ongoing phase I trial (NCT04502602) to investigate optimal dosing for the combination of neratinib and niraparib in platinum-resistant ovarian cancer.

EGFR and Met have also been implicated in modulation of PARP inhibitor sensitivity.32,98 Since multiple inhibitors of these RTKs are clinically used for treatment of non-EOC types and exhibit non-overlapping toxicities with the PARP inhibitors, combining these two classes of drugs may represent a line of rapid bench-to-bedside drug development for EOC. It is of particular attraction, since there are growing cases of clinical resistance to the PARP inhibitors in EOC patient populations.99,100

Co-inhibition with transcriptional and epigenetic mediators

Another hurdle in applying RTK inhibitors for EOC treatment is frequent activation of their downstream transcriptional and epigenetic mediators. Notably, a large portion of ovarian tumors exhibit Myc amplification.23,101 This type of clinical malignancy seems vulnerable to the targeting of BRD4, a transcriptional and epigenetic mediator and a member of the bromodomain-containing protein family, based on our study and others.22,23,102 Yet, sustained treatment with BRD4 inhibitor, JQ1, an investigational inhibitor interfering with the interaction between BRD4 and the transcription factor Myc and histone proteins, leads to an adaptive response of several RTKs.51 In addition, there is evidence that members of the BET family drive resistance to MEK inhibitors in ovarian cancer cells through activation of RTKs, notably ERBB3.56 Hence, the concomitant targeting of the RTKs and epigenetic drivers may constitute another line of promising synthetic lethal therapy for EOC treatment.

Functional roles and targeting of the non-ERBB RTKs

VEGF/VEGFR-driven pathways and angiogenesis

Thus far, the most successful clinical targeting of the RTKs in EOC has been through the monoclonal antibody-based inhibition of the VEGF-VEGFR axis.103 There is a growing consensus that tumor growth and dissemination are highly dependent on the formation of new blood vessels, a process called angiogenesis.104 In human ovarian tumors, the VEGFR2 signaling pathway is highly activated.55 This receptor drives angiogenesis in an autocrine fashion through physical interaction with its ligand, VEGF-A.104,105 In EOC, tumor angiogenesis is highly prone to inhibition by the VEGF-neutralizing monoclonal antibody, bevacizumab (Table 3).106 In addition, bevacizumab appears to have a growth-suppressive impact on EOC tumors.25 This inhibitory effect seems markedly enhanced when co-administered with chemotherapy.107,108 Intriguingly, such a drug offers a limited impact on patient survival duration.42,103 Nonetheless, bevacizumab is now widely used together with olaparib, a PARP inhibitor, as maintenance therapy for women with tumors exhibiting the BRCA mutation or genomic instability and/or who have shown response to platinum-based chemotherapy.109 In contrast, the use of bevacizumab in conjunction with the immune checkpoint inhibitors appears to have limited clinical benefit based on a recent study.42

Table 3.

A glimpse of the clinical trials with dual blockade of the VEGF/VEGFR axis and the ERBB receptors in ovarian/genitourinary cancers

Targets Intervention Clinical setting Trial ID Phase Enrollment
VEGF
& EGFR
bevacizumab + erlotinib advanced, refractory ovarian cancer NCT00130520 II 40
bevacizumab + erlotinib recurrent or metastatic ovarian epithelial, fallopian tube, or primary peritoneal cavity cancer NCT00126542 II 35
experimental: carboplatin + paclitaxel + bevacizumab then bevacizumab
experimental: carboplatin + paclitaxel + bevacizumab then bevacizumab + erlotinib
first-line treatment of newly diagnosed advanced ovarian, fallopian tube, primary peritoneal cancer and papillary serous or clear cell Mullerian tumors NCT00520013 II 60
VEGFR
& ERBB2
experimental: combination arm (pazopanib plus lapatinib)
active comparator: lapatinib monotherapy (lapatinib)
active comparator: pazopanib monotherapy (pazopanib)
recurrent or persistent advanced metastatic cervical cancer NCT00430781 II 228

c-MET and its inhibitors

Next to the ERBB family, c-Met has received the most attention regarding the clinical prognosis and therapeutic targeting in human cancers. This kinase is upregulated in 10% of the ovarian cancer cases in a previous study, as well as in our TGCA cohort (Figure 2), and it is strongly implicated in promotion of tumor progression and metastasis.110,111 Recently, c-Met has been linked to tumor resistance to the PARP inhibitors.100,112 This resistance can readily be overcome by combining the PARP inhibitor talazoparib with the multi-kinase inhibitor crizotinib.32 Mechanistically, it may involve c-Met-mediated phosphorylation of the PARP1 proteins.113 Meanwhile, the combined inhibition of c-Met and EGFR has been found to sensitize tumor cells response to alazoparib, a PARP inhibitor, in breast cancer.32,33 Consistent with this evidence, the combination of EGFR and c-Met inhibitors exhibited additive or synergistic inhibitory effects on tumor growth and metastatic potential.110,111 Conceivably, simultaneous targeting of c-Met and EGFR may provide an alternative strategy for sensitizing some EOC tumors to the PARP inhibitors.

Other RTK inhibitors

Another class of RTKs, the Eph family, has also been linked to EOC malignancy.27 EPH kinases are activated upon interaction/binding with their ligands on the surface of the opposing stroma cells (Figure 1).70 In the TCGA cohort, several members of the Eph family are upregulated (Figure 2), consistent with recent studies.27,69 There is also evidence that some members of the Eph family are dysregulated at the protein level and correlate with patient survival.27,69 To date, multiple inhibitors, including NVP-BHG712 and a class of xanthine-based chemical inhibitors, have been developed to target EPHB4 kinase in EOC.114,115.

It is worth noting that additional RTKs, such as ALK and AXL, have been found to be active or overexpressed in EOC (Figure 1).116,117 They are altered at the genomic level in 10% of EOC tumors (Figure 2). These RTKs appear to play a role in metastasis through regulation of the tumor microenvironment.118 Recently, APG-2449, a promising inhibitor of ALK kinase, has been tested for EOC targeting in a preclinical model.119 BGB324, a small-molecule inhibitor of AXL, has been shown to increase tumor sensitivity to paclitaxel and carboplatin in a PDX model-based EOC study.120

Strategies for overcoming clinical resistance to RTK targeting

Co-inhibition of RTKs and constitutively active PI3K/Akt-dependent pathways

It is increasingly clear that intrinsic or acquired resistance to ERBB2 or other RTK inhibitors across human cancer types is strongly tied to constitutive activation of their downstream effectors, particularly intermediates of the PI3K/AKT/mTOR pathway.30,38 This concept is supported by our analysis of the TCGA cohort (Figures 2 and 3). One straightforward approach to overcoming such resistance to ERBB targeting is to combine inhibitors of both ERBB2 and PI3K/AKT or mTOR kinases for EOC treatment (Figures 1 and 3). Thus far, several inhibitors of the PI3K/AKT/mTOR pathway have been evaluated for EOC in combination with the ERBB antagonists.30,121 Notably, in a recent randomized phase II trial, the combination of buparlisib, a pan-PI3K inhibitor, and trastuzumab plus paclitaxel is associated with a higher overall patient response rate as compared with the placebo arm.122 Meanwhile, the efficacy of this combinatorial targeting may be boosted by co-targeting estrogen-dependent functions/pathways, as this hormone is elevated in >60% of ovarian cancer cases and is consistently implicated in onset and progression of ovarian cancer.123,124 Conceivably, with the aid of ER-based stratification, ER+/ERBB2+ patient populations are likely more responsive to co-inhibition of ERBB2 and PI3K, and acquire the ability to overcome intrinsic resistance to the ERBB2 antagonists.

Co-inhibition of RTKs and constitutively activated RAS/MEK pathways

Another signaling pathway behind EOC resistance to ERBB targeting is the RAS/MAPK pathway (Figure 1). A number of intermediates of this oncogenic pathway have been found to be mutated and/or constitutively activated in EOC tumors, and are associated with resistance to traditional chemotherapies and rapid disease progression.56,125 In addition, several RTKs were found activated in NF1-deficient ovarian tumor cells after prolonged MEK inhibitor treatment.56 Hence, the combination of ERBB antagonists and inhibitors of the RAS-MEK pathway may represent another viable targeting option for EOC treatment.

Co-targeting of RTKs and pro-cell survival pathways

Co-inhibition of the anti-apoptotic pathways: ERBB oncogenes are also known to drive drug resistance in the clinic through activation of the anti-apoptotic pathways. This scenario is confounded by the evidence that many of the anti-apoptotic mediators, such as Bcl-2, a well-known anti-apoptotic factor, are actually dysregulated at the genomic level.126 Hence, co-targeting of this pathway and ERBB receptors could improve efficacy of the RTK antagonists in EOC.

Co-targeting the integrin/FAK axis: another pro-survival pathway utilized by cancer cells is the integrin/FAK signaling axis. This axis has long been recognized to crosstalk with multiple ERBB receptors during tumor development and progression.57 In addition, it is strongly activated in the ovarian tumor cells carrying TP53 mutations.127 Conceivably, dual blockade of the ERBB- and integrin-dependent signal transduction may synergistically hinder tumor growth. Meanwhile, FAK, a key effector of integrin signaling, is extensively amplified with the Myc oncogene in many ovarian tumors.22,23 Clinically, such genomic landscape may also represent a distinct class of EOC malignancy, as tumors with such genomic characteristics are highly refractory to traditional chemotherapies based on in vitro studies.23,128 In addition, tumor cells carrying the FAK-associated 8q24 amplicon appear more susceptible to PARP inhibition.26 In line with this notion, MRCKA has been shown to drive the activity of the integrin/FAK axis to impact EOC malignancy, presenting an alternative targeting strategy.128 This finding and the data from an evaluation with a MEK inhibitor, are consistent with our recent study on co-targeting of FAK and Myc in EOC.23,56 Overall, co-targeting of the ERBB oncogenes and the integrin/FAK axis may represent another potential therapeutic option for EOC.

Co-suppression of the inflammatory pathways: the activation of inflammatory pathways through the NF-κB-based signaling network is also implicated in tumor resistance to RTK inhibitors in ovarian cancer.54 Recently, JAK2 has been shown to promote resistance to the RTK inhibitors in EOC cells.54,129 The NF-κB-based network may also indirectly impact EOC resistance to RTK inhibitors through FAK-mediated recruitment of stromal cells, such as myeloid-derived suppressive cells.130

Perspective on therapeutic targeting of the RTKs

Owing to significant toxicities and rapid development of drug resistance to standard chemotherapy encountered during EOC treatment, recent research efforts have been directed toward development of individualized therapies. This endeavor is heightened by broad heterogeneity among EOC tumors. However, such a therapeutic strategy becomes increasingly achievable with powerful multi-omics technologies to rapidly profile key genetic alterations and activation of oncogenic pathways in the EOC patient biopsies. It will also provide rapid detection and quantification of dysregulated RTK signaling in tumor cells, mediators of resistance to the RTK inhibitors or state of re-activated RTKs, thereby fueling the RTK antagonists as a potential second or third line of therapy for EOC. In addition, the ERBB2-based antibody conjugates may represent another therapeutic option for EOC.131,132,133 This class of ERBB2 drugs is particularly appealing, as they are target specific and possess a favorable toxicity profile.134

Decades of experimental and clinical studies have accumulated evidence that any single agent-based targeting is usually insufficient for eradication of EOC, as the disease is propelled by a wide range of oncogenic dysregulation and intrinsic resistance to traditional chemotherapies. This clinical challenge is further aggravated by acquired drug resistance through influx of cancer stem cells, evolving microenvironments, and expansion of intratumoral heterogeneity.15,117,126,135,136,137,138 Despite such a complex landscape, EOC tumors sometimes appear particularly vulnerable to certain targeting. Notably, the BRCA1/2 mutated ovarian tumors are highly susceptible to inhibition of the PARP enzyme-dependent pathway, as such a pathway is essential for DNA repair and cell survival. As a result, PARP inhibitors provide a line of synthetic lethal targeting for EOC.9,139 Based on recent advances in genomic understanding of EOC, such a strategy may be enhanced by adding RTK inhibitors in the wake of overcoming adaptive therapeutic resistance.23 Finally, there is still promise for developing targeted therapies through combination of RTK antagonists and immune checkpoint inhibitors. This strategy may exert dual therapeutic impact on EOC, as some RTK-targeting agents simultaneously mediate antitumor activity of immune cells, including natural killer cells.140

Conclusions

In conclusion, the oncogenic ERBB members and other RTKs are frequently dysregulated in EOC at the genomic and expression levels. Their oncogenic signaling is strengthened by diverse genomic alterations or mutations of key intermediates of the PI3K/AKT and RAS/MAPK pathways during onset, growth, and progression of EOC. Some RTK signaling pathways are also revoked in the course of prolonged use of systemic and targeted therapeutic agents. As a result, the RTK inhibitors are well positioned as potential second- or third-line therapy for EOC treatment in the context of combinatorial targeting.

Acknowledgments

This study was supported in part by a pilot project grant (no. IRG 85-001-25) from the American Cancer Society) to X.H.Y. and 2020 USTC-Affiliated Hospital Introduction Project to Medical Leading Technology (grant no. 2020LXJS-05) to Ying Zhou.

Author contributions

Y. Wei, S.E., D.S., R.d.G., J.Q., J.S., and K.C. performed literature and database search and analyses, and wrote the manuscript. X.H.Y., F.U., B.B.Y., R.C., L.A.B., Y. Wu, T.O., B.Z., Y.Z., W.Z., and D.W. contributed to the development of the scientific concept, critical discussion on recent experimental studies, pharmacological analyses and clinical application of the RTK inhibitors, and their potential use for combinatorial targeting. These authors were all involved in the supervision of writing and editing of the manuscript. X.H.Y. and B.B.Y. are the lead contacts and coordinated the manuscript preparation and submission, and related scientific and clinical discussion.

Declaration of interests

The authors declare no competing interests.

Contributor Information

Ying Zhou, Email: caddiezy@ustc.edu.cn.

Weidong Zhao, Email: vctorzhao@ustc.edu.cn.

Burton B. Yang, Email: byang@sri.utoronto.ca.

Frederick R. Ueland, Email: fuela0@uky.edu.

Xiuwei H. Yang, Email: xiuwei-yang@uky.edu.

References

  • 1.Otsuka I. Mechanisms of high-grade serous carcinogenesis in the fallopian tube and ovary: current hypotheses, etiologic factors, and molecular alterations. Int. J. Mol. Sci. 2021;22:4409. doi: 10.3390/ijms22094409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Otsuka I., Matsuura T. Screening and prevention for high-grade serous carcinoma of the ovary based on carcinogenesis-fallopian tube- and ovarian-derived tumors and incessant retrograde bleeding. Diagnostics (Basel) 2020;10:120. doi: 10.3390/diagnostics10020120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kurnit K.C., Fleming G.F., Lengyel E. Updates and new options in advanced epithelial ovarian cancer treatment. Obstet. Gynecol. 2021;137:108–121. doi: 10.1097/AOG.0000000000004173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kim A., Ueda Y., Naka T., Enomoto T. Therapeutic strategies in epithelial ovarian cancer. J. Exp. Clin. Cancer Res. 2012;31:14. doi: 10.1186/1756-9966-31-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Colombo P.E., Fabbro M., Theillet C., Bibeau F., Rouanet P., Ray-Coquard I. Sensitivity and resistance to treatment in the primary management of epithelial ovarian cancer. Crit. Rev. Oncol. Hematol. 2014;89:207–216. doi: 10.1016/j.critrevonc.2013.08.017. [DOI] [PubMed] [Google Scholar]
  • 6.Kemp Z., Ledermann J. Update on first-line treatment of advanced ovarian carcinoma. Int. J. Womens Health. 2013;5:45–51. doi: 10.2147/IJWH.S30231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Davis A., Tinker A.V., Friedlander M. "Platinum resistant" ovarian cancer: what is it, who to treat and how to measure benefit? Gynecol. Oncol. 2014;133:624–631. doi: 10.1016/j.ygyno.2014.02.038. [DOI] [PubMed] [Google Scholar]
  • 8.Dizon D.S. PARP inhibitors for targeted treatment in ovarian cancer. Lancet. 2017;390:1929–1930. doi: 10.1016/S0140-6736(17)32418-2. [DOI] [PubMed] [Google Scholar]
  • 9.Muñoz-Galván S., Carnero A. Leveraging genomics, transcriptomics, and epigenomics to understand the biology and chemoresistance of ovarian cancer. Cancers (Basel) 2021;13:4029. doi: 10.3390/cancers13164029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Geistlinger L., Oh S., Ramos M., Schiffer L., LaRue R.S., Henzler C.M., Munro S.A., Daughters C., Nelson A.C., Winterhoff B.J., et al. Multiomic analysis of subtype evolution and heterogeneity in high-grade serous ovarian carcinoma. Cancer Res. 2020;80:4335–4345. doi: 10.1158/0008-5472.CAN-20-0521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Dugo M., Devecchi A., De Cecco L., Cecchin E., Mezzanzanica D., Sensi M., Bagnoli M. Focal recurrent copy number alterations characterize disease relapse in high grade serous ovarian cancer patients with good clinical prognosis: a pilot study. Genes (Basel) 2019;10:678. doi: 10.3390/genes10090678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dentro S.C., Leshchiner I., Haase K., Tarabichi M., Wintersinger J., Deshwar A.G., Yu K., Rubanova Y., Macintyre G., Demeulemeester J., et al. Characterizing genetic intra-tumor heterogeneity across 2,658 human cancer genomes. Cell. 2021;184:2239–2254.e39. doi: 10.1016/j.cell.2021.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Choi Y.J., Rhee J.K., Hur S.Y., Kim M.S., Lee S.H., Chung Y.J., Kim T.M., Lee S.H. Intraindividual genomic heterogeneity of high-grade serous carcinoma of the ovary and clinical utility of ascitic cancer cells for mutation profiling. J. Pathol. 2017;241:57–66. doi: 10.1002/path.4819. [DOI] [PubMed] [Google Scholar]
  • 14.Lee S., Zhao L., Rojas C., Bateman N.W., Yao H., Lara O.D., Celestino J., Morgan M.B., Nguyen T.V., Conrads K.A., et al. Molecular analysis of clinically defined subsets of high-grade serous ovarian cancer. Cell Rep. 2020;31:107502. doi: 10.1016/j.celrep.2020.03.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Abelson S., Shamai Y., Berger L., Shouval R., Skorecki K., Tzukerman M. Intratumoral heterogeneity in the self-renewal and tumorigenic differentiation of ovarian cancer. Stem Cells. 2012;30:415–424. doi: 10.1002/stem.1029. [DOI] [PubMed] [Google Scholar]
  • 16.Mazor T., Pankov A., Song J.S., Costello J.F. Intratumoral heterogeneity of the epigenome. Cancer Cell. 2016;29:440–451. doi: 10.1016/j.ccell.2016.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chen G.M., Kannan L., Geistlinger L., Kofia V., Safikhani Z., Gendoo D.M.A., Parmigiani G., Birrer M., Haibe-Kains B., Waldron L. Consensus on molecular subtypes of high-grade serous ovarian carcinoma. Clin. Cancer Res. 2018;24:5037–5047. doi: 10.1158/1078-0432.CCR-18-0784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Schwede M., Waldron L., Mok S.C., Wei W., Basunia A., Merritt M.A., Mitsiades C.S., Parmigiani G., Harrington D.P., Quackenbush J., et al. The impact of stroma admixture on molecular subtypes and prognostic gene signatures in serous ovarian cancer. Cancer Epidemiol. Biomarkers Prev. 2020;29:509–519. doi: 10.1158/1055-9965.EPI-18-1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.The Cancer Genome Atlas Research Network Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–615. doi: 10.1038/nature10166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Marcotte R., Brown K.R., Suarez F., Sayad A., Karamboulas K., Krzyzanowski P.M., Sircoulomb F., Medrano M., Fedyshyn Y., Koh J.L.Y., et al. Essential gene profiles in breast, pancreatic, and ovarian cancer cells. Cancer Discov. 2012;2:172–189. doi: 10.1158/2159-8290.CD-11-0224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Murakami R., Matsumura N., Brown J.B., Higasa K., Tsutsumi T., Kamada M., Abou-Taleb H., Hosoe Y., Kitamura S., Yamaguchi K., et al. Exome sequencing landscape analysis in ovarian clear cell carcinoma shed light on key chromosomal regions and mutation gene networks. Am. J. Pathol. 2017;187:2246–2258. doi: 10.1016/j.ajpath.2017.06.012. [DOI] [PubMed] [Google Scholar]
  • 22.Diaz Osterman C.J., Ozmadenci D., Kleinschmidt E.G., Taylor K.N., Barrie A.M., Jiang S., Bean L.M., Sulzmaier F.J., Jean C., Tancioni I., et al. FAK activity sustains intrinsic and acquired ovarian cancer resistance to platinum chemotherapy. Elife. 2019;8:e47327. doi: 10.7554/eLife.47327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Xu B., Lefringhouse J., Liu Z., West D., Baldwin L.A., Ou C., Chen L., Napier D., Chaiswing L., Brewer L.D., et al. Inhibition of the integrin/FAK signaling axis and c-Myc synergistically disrupts ovarian cancer malignancy. Oncogenesis. 2017;6:e295. doi: 10.1038/oncsis.2016.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hynes N.E., MacDonald G. ErbB receptors and signaling pathways in cancer. Curr. Opin. Cell Biol. 2009;21:177–184. doi: 10.1016/j.ceb.2008.12.010. [DOI] [PubMed] [Google Scholar]
  • 25.Tewari K.S., Burger R.A., Enserro D., Norquist B.M., Swisher E.M., Brady M.F., Bookman M.A., Fleming G.F., Huang H., Homesley H.D., et al. Final overall survival of a randomized trial of bevacizumab for primary treatment of ovarian cancer. J. Clin. Oncol. 2019;37:2317–2328. doi: 10.1200/JCO.19.01009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Papp E., Hallberg D., Konecny G.E., Bruhm D.C., Adleff V., Noë M., Kagiampakis I., Palsgrove D., Conklin D., Kinose Y., et al. Integrated genomic, epigenomic, and expression analyses of ovarian cancer cell lines. Cell Rep. 2018;25:2617–2633. doi: 10.1016/j.celrep.2018.10.096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Psilopatis I., Pergaris A., Vrettou K., Tsourouflis G., Theocharis S. The EPH/ephrin system in gynecological cancers: focusing on the roots of carcinogenesis for better patient management. Int. J. Mol. Sci. 2022;23:3249. doi: 10.3390/ijms23063249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ray-Coquard I., Pautier P., Pignata S., Pérol D., González-Martín A., Berger R., Fujiwara K., Vergote I., Colombo N., Mäenpää J., et al. Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. N. Engl. J. Med. 2019;381:2416–2428. doi: 10.1056/NEJMoa1911361. [DOI] [PubMed] [Google Scholar]
  • 29.Haunschild C.E., Tewari K.S. Bevacizumab use in the frontline, maintenance and recurrent settings for ovarian cancer. Future Oncol. 2020;16:225–246. doi: 10.2217/fon-2019-0042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Katopodis P., Chudasama D., Wander G., Sales L., Kumar J., Pandhal M., Anikin V., Chatterjee J., Hall M., Karteris E. Kinase inhibitors and ovarian cancer. Cancers (Basel) 2019;11:1357. doi: 10.3390/cancers11091357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bouhaddou M., Eckhardt M., Chi Naing Z.Z., Kim M., Ideker T., Krogan N.J. Mapping the protein-protein and genetic interactions of cancer to guide precision medicine. Curr. Opin. Genet. Dev. 2019;54:110–117. doi: 10.1016/j.gde.2019.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chu Y.Y., Yam C., Chen M.K., Chan L.C., Xiao M., Wei Y.K., Yamaguchi H., Lee P.C., Han Y., Nie L., et al. Blocking c-Met and EGFR reverses acquired resistance of PARP inhibitors in triple-negative breast cancer. Am. J. Cancer Res. 2020;10:648–661. [PMC free article] [PubMed] [Google Scholar]
  • 33.Dong Q., Du Y., Li H., Liu C., Wei Y., Chen M.K., Zhao X., Chu Y.Y., Qiu Y., Qin L., et al. EGFR and c-MET cooperate to enhance resistance to PARP inhibitors in hepatocellular carcinoma. Cancer Res. 2019;79:819–829. doi: 10.1158/0008-5472.CAN-18-1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kondrashova O., Topp M., Nesic K., Lieschke E., Ho G.Y., Harrell M.I., Zapparoli G.V., Hadley A., Holian R., Boehm E., et al. Methylation of all BRCA1 copies predicts response to the PARP inhibitor rucaparib in ovarian carcinoma. Nat. Commun. 2018;9:3970. doi: 10.1038/s41467-018-05564-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bell-McGuinn K.M., Matthews C.M., Ho S.N., Barve M., Gilbert L., Penson R.T., Lengyel E., Palaparthy R., Gilder K., Vassos A., et al. A phase II, single-arm study of the anti-alpha5beta1 integrin antibody volociximab as monotherapy in patients with platinum-resistant advanced epithelial ovarian or primary peritoneal cancer. Gynecol. Oncol. 2011;121:273–279. doi: 10.1016/j.ygyno.2010.12.362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Coleman R.L., Oza A.M., Lorusso D., Aghajanian C., Oaknin A., Dean A., Colombo N., Weberpals J.I., Clamp A., Scambia G., et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390:1949–1961. doi: 10.1016/S0140-6736(17)32440-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Swisher E.M., Lin K.K., Oza A.M., Scott C.L., Giordano H., Sun J., Konecny G.E., Coleman R.L., Tinker A.V., O'Malley D.M., et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol. 2017;18:75–87. doi: 10.1016/S1470-2045(16)30559-9. [DOI] [PubMed] [Google Scholar]
  • 38.Bhullar K.S., Lagarón N.O., McGowan E.M., Parmar I., Jha A., Hubbard B.P., Rupasinghe H.P.V. Kinase-targeted cancer therapies: progress, challenges and future directions. Mol. Cancer. 2018;17:48. doi: 10.1186/s12943-018-0804-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bauer K., Berger D., Zielinski C.C., Valent P., Grunt T.W. Hitting two oncogenic machineries in cancer cells: cooperative effects of the multi-kinase inhibitor ponatinib and the BET bromodomain blockers JQ1 or dBET1 on human carcinoma cells. Oncotarget. 2018;9:26491–26506. doi: 10.18632/oncotarget.25474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kurimchak A.M., Shelton C., Duncan K.E., Johnson K.J., Brown J., O'Brien S., Gabbasov R., Fink L.S., Li Y., Lounsbury N., et al. Resistance to BET bromodomain inhibitors is mediated by kinome reprogramming in ovarian cancer. Cell Rep. 2016;16:1273–1286. doi: 10.1016/j.celrep.2016.06.091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Monk B.J., Coleman R.L., Fujiwara K., Wilson M.K., Oza A.M., Oaknin A., O'Malley D.M., Lorusso D., Westin S.N., Safra T., et al. ATHENA (GOG-3020/ENGOT-ov45): a randomized, phase III trial to evaluate rucaparib as monotherapy (ATHENA-MONO) and rucaparib in combination with nivolumab (ATHENA-COMBO) as maintenance treatment following frontline platinum-based chemotherapy in ovarian cancer. Int. J. Gynecol. Cancer. 2021;31:1589–1594. doi: 10.1136/ijgc-2021-002933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Moore K.N., Bookman M., Sehouli J., Miller A., Anderson C., Scambia G., Myers T., Taskiran C., Robison K., Mäenpää J., et al. Atezolizumab, bevacizumab, and chemotherapy for newly diagnosed stage III or IV ovarian cancer: placebo-controlled randomized phase III trial (IMagyn050/GOG 3015/ENGOT-OV39) J. Clin. Oncol. 2021;39:1842–1855. doi: 10.1200/JCO.21.00306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Baselga J., Swain S.M. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat. Rev. Cancer. 2009;9:463–475. doi: 10.1038/nrc2656. [DOI] [PubMed] [Google Scholar]
  • 44.Casaletto J.B., McClatchey A.I. Spatial regulation of receptor tyrosine kinases in development and cancer. Nat. Rev. Cancer. 2012;12:387–400. doi: 10.1038/nrc3277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Chiasson-MacKenzie C., McClatchey A.I. Cell-cell contact and receptor tyrosine kinase signaling. Cold Spring Harb. Perspect. Biol. 2018;10:a029215. doi: 10.1101/cshperspect.a029215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Shi W., Fan H., Shum L., Derynck R. The tetraspanin CD9 associates with transmembrane TGF-alpha and regulates TGF-alpha-induced EGF receptor activation and cell proliferation. J. Cell Biol. 2000;148:591–602. doi: 10.1083/jcb.148.3.591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ruiz-Saenz A., Moasser M.M. Targeting HER2 by combination therapies. J. Clin. Oncol. 2018;36:808–811. doi: 10.1200/JCO.2017.77.1899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Tebbutt N., Pedersen M.W., Johns T.G. Targeting the ERBB family in cancer: couples therapy. Nat. Rev. Cancer. 2013;13:663–673. doi: 10.1038/nrc3559. [DOI] [PubMed] [Google Scholar]
  • 49.Yarden Y., Pines G. The ERBB network: at last, cancer therapy meets systems biology. Nat. Rev. Cancer. 2012;12:553–563. doi: 10.1038/nrc3309. [DOI] [PubMed] [Google Scholar]
  • 50.Ferguson K.M. Structure-based view of epidermal growth factor receptor regulation. Annu. Rev. Biophys. 2008;37:353–373. doi: 10.1146/annurev.biophys.37.032807.125829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Liu F., Hon G.C., Villa G.R., Turner K.M., Ikegami S., Yang H., Ye Z., Li B., Kuan S., Lee A.Y., et al. EGFR mutation promotes glioblastoma through epigenome and transcription factor network remodeling. Mol. Cell. 2015;60:307–318. doi: 10.1016/j.molcel.2015.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Yang X.H., Flores L.M., Li Q., Zhou P., Xu F., Krop I.E., Hemler M.E. Disruption of laminin-integrin-CD151-focal adhesion kinase axis sensitizes breast cancer cells to ErbB2 antagonists. Cancer Res. 2010;70:2256–2263. doi: 10.1158/0008-5472.CAN-09-4032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Pane K., Affinito O., Zanfardino M., Castaldo R., Incoronato M., Salvatore M., Franzese M. An integrative computational approach based on expression similarity signatures to identify protein-protein interaction networks in Female-specific cancers. Front. Genet. 2020;11:612521. doi: 10.3389/fgene.2020.612521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wen W., Wu J., Liu L., Tian Y., Buettner R., Hsieh M.Y., Horne D., Dellinger T.H., Han E.S., Jove R., Yim J.H. Synergistic anti-tumor effect of combined inhibition of EGFR and JAK/STAT3 pathways in human ovarian cancer. Mol. Cancer. 2015;14:100. doi: 10.1186/s12943-015-0366-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Trinh X.B., Tjalma W.A.A., Vermeulen P.B., Van den Eynden G., Van der Auwera I., Van Laere S.J., Helleman J., Berns E.M., Dirix L.Y., van Dam P.A. The VEGF pathway and the AKT/mTOR/p70S6K1 signalling pathway in human epithelial ovarian cancer. Br. J. Cancer. 2009;100:971–978. doi: 10.1038/sj.bjc.6604921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kurimchak A.M., Shelton C., Herrera-Montávez C., Duncan K.E., Chernoff J., Duncan J.S. Intrinsic resistance to MEK inhibition through BET protein-mediated kinome reprogramming in NF1-deficient ovarian cancer. Mol. Cancer Res. 2019;17:1721–1734. doi: 10.1158/1541-7786.MCR-18-1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Cooper J., Giancotti F.G. Integrin signaling in cancer: mechanotransduction, stemness, epithelial plasticity, and therapeutic resistance. Cancer Cell. 2019;35:347–367. doi: 10.1016/j.ccell.2019.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Teplinsky E., Muggia F. Targeting HER2 in ovarian and uterine cancers: challenges and future directions. Gynecol. Oncol. 2014;135:364–370. doi: 10.1016/j.ygyno.2014.09.003. [DOI] [PubMed] [Google Scholar]
  • 59.Wang Z. ErbB receptors and cancer. Methods Mol. Biol. 2017;1652:3–35. doi: 10.1007/978-1-4939-7219-7_1. [DOI] [PubMed] [Google Scholar]
  • 60.Gui T., Shen K. The epidermal growth factor receptor as a therapeutic target in epithelial ovarian cancer. Cancer Epidemiol. 2012;36:490–496. doi: 10.1016/j.canep.2012.06.005. [DOI] [PubMed] [Google Scholar]
  • 61.Psyrri A., Kassar M., Yu Z., Bamias A., Weinberger P.M., Markakis S., Kowalski D., Camp R.L., Rimm D.L., Dimopoulos M.A. Effect of epidermal growth factor receptor expression level on survival in patients with epithelial ovarian cancer. Clin. Cancer Res. 2005;11:8637–8643. doi: 10.1158/1078-0432.CCR-05-1436. [DOI] [PubMed] [Google Scholar]
  • 62.Serrano-Olvera A., Dueñas-González A., Gallardo-Rincón D., Candelaria M., De la Garza-Salazar J. Prognostic, predictive and therapeutic implications of HER2 in invasive epithelial ovarian cancer. Cancer Treat. Rev. 2006;32:180–190. doi: 10.1016/j.ctrv.2006.01.001. [DOI] [PubMed] [Google Scholar]
  • 63.Reibenwein J., Krainer M. Targeting signaling pathways in ovarian cancer. Expert Opin. Ther. Targets. 2008;12:353–365. doi: 10.1517/14728222.12.3.353. [DOI] [PubMed] [Google Scholar]
  • 64.Luo H., Xu X., Ye M., Sheng B., Zhu X. The prognostic value of HER2 in ovarian cancer: a meta-analysis of observational studies. PLoS one. 2018;13:e0191972. doi: 10.1371/journal.pone.0191972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.van der Horst E.H., Murgia M., Treder M., Ullrich A. Anti-HER-3 MAbs inhibit HER-3-mediated signaling in breast cancer cell lines resistant to anti-HER-2 antibodies. Int. J. Cancer. 2005;115:519–527. doi: 10.1002/ijc.20867. [DOI] [PubMed] [Google Scholar]
  • 66.Chung Y.W., Kim S., Hong J.H., Lee J.K., Lee N.W., Lee Y.S., Song J.Y. Overexpression of HER2/HER3 and clinical feature of ovarian cancer. J. Gynecol. Oncol. 2019;30:e75. doi: 10.3802/jgo.2019.30.e75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Li H., Zeng J., Shen K. PI3K/AKT/mTOR signaling pathway as a therapeutic target for ovarian cancer. Arch. Gynecol. Obstet. 2014;290:1067–1078. doi: 10.1007/s00404-014-3377-3. [DOI] [PubMed] [Google Scholar]
  • 68.Gasparri M.L., Besharat Z.M., Farooqi A.A., Khalid S., Taghavi K., Besharat R.A., Sabato C., Papadia A., Panici P.B., Mueller M.D., Ferretti E. MiRNAs and their interplay with PI3K/AKT/mTOR pathway in ovarian cancer cells: a potential role in platinum resistance. J. Cancer Res. Clin. Oncol. 2018;144:2313–2318. doi: 10.1007/s00432-018-2737-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Jukonen J., Moyano-Galceran L., Höpfner K., Pietilä E.A., Lehtinen L., Huhtinen K., Gucciardo E., Hynninen J., Hietanen S., Grénman S., et al. Aggressive and recurrent ovarian cancers upregulate ephrinA5, a non-canonical effector of EphA2 signaling duality. Sci. Rep. 2021;11:8856. doi: 10.1038/s41598-021-88382-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Kania A., Klein R. Mechanisms of ephrin-Eph signalling in development, physiology and disease. Nat. Rev. Mol. Cell Biol. 2016;17:240–256. doi: 10.1038/nrm.2015.16. [DOI] [PubMed] [Google Scholar]
  • 71.Hirte H.W. Profile of erlotinib and its potential in the treatment of advanced ovarian carcinoma. Onco. Targets Ther. 2013;6:427–435. doi: 10.2147/OTT.S30373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Barlaam B., Acton D.G., Ballard P., Bradbury R.H., Cross D., Ducray R., Germain H., Hudson K., Klinowska T., Magnien F., et al. Neutral 5-substituted 4-indazolylaminoquinazolines as potent, orally active inhibitors of erbB2 receptor tyrosine kinase. Bioorg. Med. Chem. Lett. 2008;18:1799–1803. doi: 10.1016/j.bmcl.2008.02.035. [DOI] [PubMed] [Google Scholar]
  • 73.McCorkle J.R., Gorski J.W., Liu J., Riggs M.B., McDowell A.B., Lin N., Wang C., Ueland F.R., Kolesar J.M. Lapatinib and poziotinib overcome ABCB1-mediated paclitaxel resistance in ovarian cancer. PLoS One. 2021;16:e0254205. doi: 10.1371/journal.pone.0254205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Diz Taín P., González A.L., García-Palomo A. [Mechanism of action and preclinical development of afatinib] Med. Clin. 2016;146:7–11. doi: 10.1016/S0025-7753(16)30257-3. [DOI] [PubMed] [Google Scholar]
  • 75.Mehner C., Oberg A.L., Goergen K.M., Kalli K.R., Maurer M.J., Nassar A., Goode E.L., Keeney G.L., Jatoi A., Radisky D.C., Radisky E.S. EGFR as a prognostic biomarker and therapeutic target in ovarian cancer: evaluation of patient cohort and literature review. Genes Cancer. 2017;8:589–599. doi: 10.18632/genesandcancer.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Schwab C.L., Bellone S., English D.P., Roque D.M., Lopez S., Cocco E., Nicoletti R., Bortolomai I., Bonazzoli E., Ratner E., et al. Afatinib demonstrates remarkable activity against HER2-amplified uterine serous endometrial cancer in vitro and in vivo. Br. J. Cancer. 2014;111:1750–1756. doi: 10.1038/bjc.2014.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Shepherd-Littlejohn A.L., Hanft W.J., Kennedy V.A., Alvarez E.A. Afatinib use in recurrent epithelial ovarian carcinoma. Gynecol. Oncol. Rep. 2019;29:70–72. doi: 10.1016/j.gore.2019.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.De Grève J., Teugels E., Geers C., Decoster L., Galdermans D., De Mey J., Everaert H., Umelo I., In't Veld P., Schallier D. Clinical activity of afatinib (BIBW 2992) in patients with lung adenocarcinoma with mutations in the kinase domain of HER2/neu. Lung Cancer. 2012;76:123–127. doi: 10.1016/j.lungcan.2012.01.008. [DOI] [PubMed] [Google Scholar]
  • 79.Garcia A.A., Sill M.W., Lankes H.A., Godwin A.K., Mannel R.S., Armstrong D.K., Carolla R.L., Liepman M.K., Spirtos N.M., Fischer E.G., Leslie K.K. A phase II evaluation of lapatinib in the treatment of persistent or recurrent epithelial ovarian or primary peritoneal carcinoma: a gynecologic oncology group study. Gynecol. Oncol. 2012;124:569–574. doi: 10.1016/j.ygyno.2011.10.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Aigner A., Hsieh S.S., Malerczyk C., Czubayko F.J.T. Reversal of HER-2 over-expression renders human ovarian cancer cells highly resistant to taxol. Toxicology. 2000;144:221–228. doi: 10.1016/s0300-483x(99)00210-3. [DOI] [PubMed] [Google Scholar]
  • 81.Abuharbeid S., Apel J., Zugmaier G., Knabbe C., Sander M., Gilbert S., Czubayko F., Aigner A. Inhibition of HER-2 by three independent targeting strategies increases paclitaxel resistance of SKOV-3 ovarian carcinoma cells. Naunyn Schmiedebergs Arch. Pharmacol. 2005;371:141. doi: 10.1007/s00210-004-1016-4. [DOI] [PubMed] [Google Scholar]
  • 82.Delord J.P., Allal C., Canal M., Mery E., Rochaix P., Hennebelle I., Pradines A., Chatelut E., Bugat R., Guichard S., Canal P. Selective inhibition of HER2 inhibits AKT signal transduction and prolongs disease-free survival in a micrometastasis model of ovarian carcinoma. Ann. Oncol. 2005;16:1889–1897. doi: 10.1093/annonc/mdi405. [DOI] [PubMed] [Google Scholar]
  • 83.Frederick P.J., Straughn J.M., Alvarez R.D., Buchsbaum D.J. Preclinical studies and clinical utilization of monoclonal antibodies in epithelial ovarian cancer. Gynecol. Oncol. 2009;113:384–390. doi: 10.1016/j.ygyno.2009.01.008. [DOI] [PubMed] [Google Scholar]
  • 84.Mullen P., Cameron D.A., Hasmann M., Smyth J.F., Langdon S.P. Sensitivity to pertuzumab (2C4) in ovarian cancer models: cross-talk with estrogen receptor signaling. Mol. Cancer Ther. 2007;6:93–100. doi: 10.1158/1535-7163.MCT-06-0401. [DOI] [PubMed] [Google Scholar]
  • 85.Gottschalk N., Kimmig R., Lang S., Singh M., Brandau S. Anti-epidermal growth factor receptor (EGFR) antibodies overcome resistance of ovarian cancer cells to targeted therapy and natural cytotoxicity. Int. J. Mol. Sci. 2012;13:12000–12016. doi: 10.3390/ijms130912000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Hirte H., Oza A., Swenerton K., Ellard S.L., Grimshaw R., Fisher B., Tsao M., Seymour L. A phase II study of erlotinib (OSI-774) given in combination with carboplatin in patients with recurrent epithelial ovarian cancer (NCIC CTG IND.149) Gynecol. Oncol. 2010;118:308–312. doi: 10.1016/j.ygyno.2010.05.005. [DOI] [PubMed] [Google Scholar]
  • 87.Despierre E., Vergote I., Anderson R., Coens C., Katsaros D., Hirsch F.R., Boeckx B., Varella-Garcia M., Ferrero A., Ray-Coquard I., et al. Epidermal growth factor receptor (EGFR) pathway biomarkers in the randomized phase III trial of erlotinib versus observation in ovarian cancer patients with No evidence of disease progression after first-line platinum-based chemotherapy. Target. Oncol. 2015;10:583–596. doi: 10.1007/s11523-015-0369-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Vergote I.B., Jimeno A., Joly F., Katsaros D., Coens C., Despierre E., Marth C., Hall M., Steer C.B., Colombo N., et al. Randomized phase III study of erlotinib versus observation in patients with no evidence of disease progression after first-line platin-based chemotherapy for ovarian carcinoma: a European Organisation for Research and Treatment of Cancer-Gynaecological Cancer Group, and Gynecologic Cancer Intergroup study. J. Clin. Oncol. 2014;32:320–326. doi: 10.1200/JCO.2013.50.5669. [DOI] [PubMed] [Google Scholar]
  • 89.Morrison J., Thoma C., Goodall R.J., Lyons T.J., Gaitskell K., Wiggans A.J., Bryant A. Epidermal growth factor receptor blockers for the treatment of ovarian cancer. Cochrane Database Syst. Rev. 2018;10:CD007927. doi: 10.1002/14651858.CD007927.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kurzeder C., Bover I., Marmé F., Rau J., Pautier P., Colombo N., Lorusso D., Ottevanger P., Bjurberg M., Marth C., et al. Double-Blind, placebo-controlled, randomized phase III trial evaluating pertuzumab combined with chemotherapy for low tumor human epidermal growth factor receptor 3 mRNA-expressing platinum-resistant ovarian cancer (PENELOPE) J. Clin. Oncol. 2016;34:2516–2525. doi: 10.1200/JCO.2015.66.0787. [DOI] [PubMed] [Google Scholar]
  • 91.Baron A.T., Wilken J.A., Haggstrom D.E., Goodrich S.T., Maihle N.J. Clinical implementation of soluble EGFR (sEGFR) as a theragnostic serum biomarker of breast, lung and ovarian cancer. IDrugs. 2009;12:302–308. [PubMed] [Google Scholar]
  • 92.Canonici A., Ivers L., Conlon N.T., Pedersen K., Gaynor N., Browne B.C., O’Brien N.A., Gullo G., Collins D.M., O’Donovan N., et al. Investigational New Drugs; 2018. HER-Targeted Tyrosine Kinase Inhibitors Enhance Response to Trastuzumab and Pertuzumab in HER2-Positive Breast Cancer. [DOI] [PubMed] [Google Scholar]
  • 93.Gagliato D.M., Jardim D.L., Marchesi M.S., Hortobagyi G.N. Mechanisms of resistance and sensitivity to anti-HER2 therapies in HER2+ breast cancer. Oncotarget. 2016;7:64431–64446. doi: 10.18632/oncotarget.7043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Chan A., Delaloge S., Holmes F.A., Moy B., Iwata H., Harvey V.J., Robert N.J., Silovski T., Gokmen E., von Minckwitz G., et al. Neratinib after trastuzumab-based adjuvant therapy in patients with HER2-positive breast cancer (ExteNET): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2016;17:367–377. doi: 10.1016/S1470-2045(15)00551-3. [DOI] [PubMed] [Google Scholar]
  • 95.Saura C., Oliveira M., Feng Y.H., Dai M.S., Chen S.W., Hurvitz S.A., Kim S.B., Moy B., Delaloge S., Gradishar W., et al. Neratinib plus capecitabine versus lapatinib plus capecitabine in HER2-positive metastatic breast cancer previously treated with >/= 2 HER2-directed regimens: phase III NALA trial. J. Clin. Oncol. 2020;38:3138–3149. doi: 10.1200/JCO.20.00147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Booth L., Roberts J.L., Samuel P., Avogadri-Connors F., Cutler R.E., Lalani A.S., Poklepovic A., Dent P. The irreversible ERBB1/2/4 inhibitor neratinib interacts with the PARP1 inhibitor niraparib to kill ovarian cancer cells. Cancer Biol. Ther. 2018;19:525–533. doi: 10.1080/15384047.2018.1436024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Booth L., Roberts J.L., Poklepovic A., Kirkwood J., Sander C., Avogadri-Connors F., Cutler R.E., Jr., Lalani A.S., Dent P. The levels of mutant K-RAS and mutant N-RAS are rapidly reduced in a Beclin1/ATG5 -dependent fashion by the irreversible ERBB1/2/4 inhibitor neratinib. Cancer Biol. Ther. 2018;19:132–137. doi: 10.1080/15384047.2017.1394556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Marcar L., Bardhan K., Gheorghiu L., Dinkelborg P., Pfäffle H., Liu Q., Wang M., Piotrowska Z., Sequist L.V., Borgmann K., et al. Acquired resistance of EGFR-mutated lung cancer to tyrosine kinase inhibitor treatment promotes PARP inhibitor sensitivity. Cell Rep. 2019;27:3422–3432.e4. doi: 10.1016/j.celrep.2019.05.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Freimund A.E., Beach J.A., Christie E.L., Bowtell D.D.L. Mechanisms of drug resistance in high-grade serous ovarian cancer. Hematol. Oncol. Clin. North Am. 2018;32:983–996. doi: 10.1016/j.hoc.2018.07.007. [DOI] [PubMed] [Google Scholar]
  • 100.Patel M., Nowsheen S., Maraboyina S., Xia F. The role of poly(ADP-ribose) polymerase inhibitors in the treatment of cancer and methods to overcome resistance: a review. Cell Biosci. 2020;10:35. doi: 10.1186/s13578-020-00390-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Phelan C.M., Kuchenbaecker K.B., Tyrer J.P., Kar S.P., Lawrenson K., Winham S.J., Dennis J., Pirie A., Riggan M.J., Chornokur G., et al. Identification of 12 new susceptibility loci for different histotypes of epithelial ovarian cancer. Nat. Genet. 2017;49:680–691. doi: 10.1038/ng.3826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Wagner R., Stübiger G., Veigel D., Wuczkowski M., Lanzerstorfer P., Weghuber J., Karteris E., Nowikovsky K., Wilfinger-Lutz N., Singer C.F., et al. Multi-level suppression of receptor-PI3K-mTORC1 by fatty acid synthase inhibitors is crucial for their efficacy against ovarian cancer cells. Oncotarget. 2017;8:11600–11613. doi: 10.18632/oncotarget.14591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Garcia J., Hurwitz H.I., Sandler A.B., Miles D., Coleman R.L., Deurloo R., Chinot O.L. Bevacizumab (Avastin(R)) in cancer treatment: a review of 15 years of clinical experience and future outlook. Cancer Treat. Rev. 2020;86:102017. doi: 10.1016/j.ctrv.2020.102017. [DOI] [PubMed] [Google Scholar]
  • 104.Lee S., Chen T.T., Barber C.L., Jordan M.C., Murdock J., Desai S., Ferrara N., Nagy A., Roos K.P., Iruela-Arispe M.L. Autocrine VEGF signaling is required for vascular homeostasis. Cell. 2007;130:691–703. doi: 10.1016/j.cell.2007.06.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kälin R.E., Kretz M.P., Meyer A.M., Kispert A., Heppner F.L., Brändli A.W. Paracrine and autocrine mechanisms of apelin signaling govern embryonic and tumor angiogenesis. Dev. Biol. 2007;305:599–614. doi: 10.1016/j.ydbio.2007.03.004. [DOI] [PubMed] [Google Scholar]
  • 106.Verheul H.M.W., Lolkema M.P.J., Qian D.Z., Hilkes Y.H.A., Liapi E., Akkerman J.W.N., Pili R., Voest E.E. Platelets take up the monoclonal antibody bevacizumab. Clin. Cancer Res. 2007;13:5341–5347. doi: 10.1158/1078-0432.CCR-07-0847. [DOI] [PubMed] [Google Scholar]
  • 107.Garcia Garcia Y., de Juan Ferré A., Mendiola C., Barretina-Ginesta M.P., Gaba Garcia L., Santaballa Bertrán A., Bover Barcelo I., Gil-Martin M., Manzano A., Rubio Pérez M.J., et al. Efficacy and safety results from GEICO 1205, a randomized phase II trial of neoadjuvant chemotherapy with or without bevacizumab for advanced epithelial ovarian cancer. Int. J. Gynecol. Cancer. 2019;29:1050–1056. doi: 10.1136/ijgc-2019-000256. [DOI] [PubMed] [Google Scholar]
  • 108.Schultheis A.M., Lurje G., Rhodes K.E., Zhang W., Yang D., Garcia A.A., Morgan R., Gandara D., Scudder S., Oza A., et al. Polymorphisms and clinical outcome in recurrent ovarian cancer treated with cyclophosphamide and bevacizumab. Clin. Cancer Res. 2008;14:7554–7563. doi: 10.1158/1078-0432.CCR-08-0351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Ruscito I., Bellati F., Ray-Coquard I., Mirza M.R., du Bois A., Gasparri M.L., Costanzi F., De Marco M.P., Nuti M., Caserta D., et al. Incorporating parp-inhibitors in primary and recurrent ovarian cancer: a meta-analysis of 12 phase II/III randomized controlled trials. Cancer Treat. Rev. 2020;87:102040. doi: 10.1016/j.ctrv.2020.102040. [DOI] [PubMed] [Google Scholar]
  • 110.Zillhardt M., Christensen J.G., Lengyel E. An orally available small-molecule inhibitor of c-Met, PF-2341066, reduces tumor burden and metastasis in a preclinical model of ovarian cancer metastasis. Neoplasia. 2010;12:1–10. doi: 10.1593/neo.09948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Sawada K., Radjabi A.R., Shinomiya N., Kistner E., Kenny H., Becker A.R., Turkyilmaz M.A., Salgia R., Yamada S.D., Vande Woude G.F., et al. c-Met overexpression is a prognostic factor in ovarian cancer and an effective target for inhibition of peritoneal dissemination and invasion. Cancer Res. 2007;67:1670–1679. doi: 10.1158/0008-5472.CAN-06-1147. [DOI] [PubMed] [Google Scholar]
  • 112.Bitler B.G., Watson Z.L., Wheeler L.J., Behbakht K. PARP inhibitors: clinical utility and possibilities of overcoming resistance. Gynecol. Oncol. 2017;147:695–704. doi: 10.1016/j.ygyno.2017.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Du Y., Yamaguchi H., Wei Y., Hsu J.L., Wang H.-L., Hsu Y.-H., Lin W.-C., Yu W.-H., Leonard P.G., Lee G.R., et al. Blocking c-Met–mediated PARP1 phosphorylation enhances anti-tumor effects of PARP inhibitors. Nat. Med. 2016;22:194–201. doi: 10.1038/nm.4032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Unzue A., Jessen-Trefzer C., Spiliotopoulos D., Gaudio E., Tarantelli C., Dong J., Zhao H., Pachmayr J., Zahler S., Bernasconi E., et al. Understanding the mechanism of action of pyrrolo[3,2-b]quinoxaline-derivatives as kinase inhibitors. RSC Med. Chem. 2020;11:665–675. doi: 10.1039/d0md00049c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Unzue A., Lafleur K., Zhao H., Zhou T., Dong J., Kolb P., Liebl J., Zahler S., Caflisch A., Nevado C. Three stories on Eph kinase inhibitors: from in silico discovery to in vivo validation. Eur. J. Med. Chem. 2016;112:347–366. doi: 10.1016/j.ejmech.2016.01.057. [DOI] [PubMed] [Google Scholar]
  • 116.Umemura S., Sowa Y., Iizumi Y., Kitawaki J., Sakai T. Synergistic effect of the inhibitors of RAF/MEK and AXL on KRAS-mutated ovarian cancer cells with high AXL expression. Cancer Sci. 2020;111:2052–2061. doi: 10.1111/cas.14414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Puvanenthiran S., Essapen S., Seddon A.M., Modjtahedi H. Impact of the putative cancer stem cell markers and growth factor receptor expression on the sensitivity of ovarian cancer cells to treatment with various forms of small molecule tyrosine kinase inhibitors and cytotoxic drugs. Int. J. Oncol. 2016;49:1825–1838. doi: 10.3892/ijo.2016.3678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Spiegel A., Brooks M.W., Houshyar S., Reinhardt F., Ardolino M., Fessler E., Chen M.B., Krall J.A., DeCock J., Zervantonakis I.K., et al. Neutrophils suppress intraluminal NK cell-mediated tumor cell clearance and enhance extravasation of disseminated carcinoma cells. Cancer Discov. 2016;6:630–649. doi: 10.1158/2159-8290.CD-15-1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Fang D.D., Tao R., Wang G., Li Y., Zhang K., Xu C., Zhai G., Wang Q., Wang J., Tang C., et al. Discovery of a novel ALK/ROS1/FAK inhibitor, APG-2449, in preclinical non-small cell lung cancer and ovarian cancer models. BMC Cancer. 2022;22:752. doi: 10.1186/s12885-022-09799-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Quinn J.M., Greenwade M.M., Palisoul M.L., Opara G., Massad K., Guo L., Zhao P., Beck-Noia H., Hagemann I.S., Hagemann A.R., et al. Therapeutic inhibition of the receptor tyrosine kinase AXL improves sensitivity to platinum and taxane in ovarian cancer. Mol. Cancer Ther. 2019;18:389–398. doi: 10.1158/1535-7163.MCT-18-0537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Keegan N.M., Gleeson J.P., Hennessy B.T., Morris P.G. PI3K inhibition to overcome endocrine resistance in breast cancer. Expert Opin. Investig. Drugs. 2018;27:1–15. doi: 10.1080/13543784.2018.1417384. [DOI] [PubMed] [Google Scholar]
  • 122.Loibl S., de la Pena L., Nekljudova V., Zardavas D., Michiels S., Denkert C., Rezai M., Bermejo B., Untch M., Lee S.C., et al. Neoadjuvant buparlisib plus trastuzumab and paclitaxel for women with HER2+ primary breast cancer: a randomised, double-blind, placebo-controlled phase II trial (NeoPHOEBE) Eur. J. Cancer. 2017;85:133–145. doi: 10.1016/j.ejca.2017.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Simpkins F., Garcia-Soto A., Slingerland J. New insights on the role of hormonal therapy in ovarian cancer. Steroids. 2013;78:530–537. doi: 10.1016/j.steroids.2013.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Wang Y., Tan S., Pan E., Ma Y., Wu X., Yu Z., Jiang K. An effective hormonal therapy for a patient with estrogen receptor 1 (ESR1)-Amplified metastatic ovarian cancer: a case report. Onco. Targets Ther. 2022;15:643–649. doi: 10.2147/OTT.S363856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Shrestha R., Llaurado Fernandez M., Dawson A., Hoenisch J., Volik S., Lin Y.Y., Anderson S., Kim H., Haegert A.M., Colborne S., et al. Multiomics characterization of low-grade serous ovarian carcinoma identifies potential biomarkers of MEK inhibitor sensitivity and therapeutic vulnerability. Cancer Res. 2021;81:1681–1694. doi: 10.1158/0008-5472.CAN-20-2222. [DOI] [PubMed] [Google Scholar]
  • 126.Momeny M., Zarrinrad G., Moghaddaskho F., Poursheikhani A., Sankanian G., Zaghal A., Mirshahvaladi S., Esmaeili F., Eyvani H., Barghi F., et al. Dacomitinib, a pan-inhibitor of ErbB receptors, suppresses growth and invasive capacity of chemoresistant ovarian carcinoma cells. Sci. Rep. 2017;7:4204. doi: 10.1038/s41598-017-04147-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Iwanicki M.P., Chen H.Y., Iavarone C., Zervantonakis I.K., Muranen T., Novak M., Ince T.A., Drapkin R., Brugge J.S. Mutant p53 regulates ovarian cancer transformed phenotypes through autocrine matrix deposition. JCI Insight. 2016;1:e86829. doi: 10.1172/jci.insight.86829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Kurimchak A.M., Herrera-Montávez C., Brown J., Johnson K.J., Sodi V., Srivastava N., Kumar V., Deihimi S., O'Brien S., Peri S., et al. Functional proteomics interrogation of the kinome identifies MRCKA as a therapeutic target in high-grade serous ovarian carcinoma. Sci. Signal. 2020;13:eaax8238. doi: 10.1126/scisignal.aax8238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Wen W., Han E.S., Dellinger T.H., Wu J., Guo Y., Buettner R., Horne D.A., Jove R., Yim J.H. Increasing antitumor activity of JAK inhibitor by simultaneous blocking multiple survival signaling pathways in human ovarian cancer. Transl. Oncol. 2019;12:1015–1025. doi: 10.1016/j.tranon.2019.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Jiang H., Hegde S., Knolhoff B.L., Zhu Y., Herndon J.M., Meyer M.A., Nywening T.M., Hawkins W.G., Shapiro I.M., Weaver D.T., et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat. Med. 2016;22:851–860. doi: 10.1038/nm.4123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Doi T., Shitara K., Naito Y., Shimomura A., Fujiwara Y., Yonemori K., Shimizu C., Shimoi T., Kuboki Y., Matsubara N., et al. Safety, pharmacokinetics, and antitumour activity of trastuzumab deruxtecan (DS-8201), a HER2-targeting antibody-drug conjugate, in patients with advanced breast and gastric or gastro-oesophageal tumours: a phase 1 dose-escalation study. Lancet Oncol. 2017;18:1512–1522. doi: 10.1016/S1470-2045(17)30604-6. [DOI] [PubMed] [Google Scholar]
  • 132.Giannone G., Montemurro F. A new player in the treatment of HER2-positive tumours. Lancet Oncol. 2019;20:748–750. doi: 10.1016/S1470-2045(19)30168-8. [DOI] [PubMed] [Google Scholar]
  • 133.Tamura K., Tsurutani J., Takahashi S., Iwata H., Krop I.E., Redfern C., Sagara Y., Doi T., Park H., Murthy R.K., et al. Trastuzumab deruxtecan (DS-8201a) in patients with advanced HER2-positive breast cancer previously treated with trastuzumab emtansine: a dose-expansion, phase 1 study. Lancet Oncol. 2019;20:816–826. doi: 10.1016/S1470-2045(19)30097-X. [DOI] [PubMed] [Google Scholar]
  • 134.Xu Z., Guo D., Jiang Z., Tong R., Jiang P., Bai L., Chen L., Zhu Y., Guo C., Shi J., Yu D. Novel HER2-targeting antibody-drug conjugates of trastuzumab beyond T-DM1 in breast cancer: trastuzumab deruxtecan(DS-8201a) and (Vic-)Trastuzumab duocarmazine (SYD985) Eur. J. Med. Chem. 2019;183:111682. doi: 10.1016/j.ejmech.2019.111682. [DOI] [PubMed] [Google Scholar]
  • 135.Schwarz R.F., Ng C.K.Y., Cooke S.L., Newman S., Temple J., Piskorz A.M., Gale D., Sayal K., Murtaza M., Baldwin P.J., et al. Spatial and temporal heterogeneity in high-grade serous ovarian cancer: a phylogenetic analysis. Plos Med. 2015;12:e1001789. doi: 10.1371/journal.pmed.1001789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Penner-Goeke S., Lichtensztejn Z., Neufeld M., Ali J.L., Altman A.D., Nachtigal M.W., McManus K.J. The temporal dynamics of chromosome instability in ovarian cancer cell lines and primary patient samples. Plos Genet. 2017;13:e1006707. doi: 10.1371/journal.pgen.1006707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Yu M., Chen S., Hong W., Gu Y., Huang B., Lin Y., Zhou Y., Jin H., Deng Y., Tu L., et al. Prognostic role of glycolysis for cancer outcome: evidence from 86 studies. J. Cancer Res. Clin. Oncol. 2019;145:967–999. doi: 10.1007/s00432-019-02847-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Liu S., Ginestier C., Charafe-Jauffret E., Foco H., Kleer C.G., Merajver S.D., Dontu G., Wicha M.S. BRCA1 regulates human mammary stem/progenitor cell fate. Proc. Natl. Acad. Sci. USA. 2008;105:1680–1685. doi: 10.1073/pnas.0711613105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Block K.I., Gyllenhaal C., Lowe L., Amedei A., Amin A., Amin A., Aquilano K., Arbiser J., Arreola A., Arzumanyan A., et al. Designing a broad-spectrum integrative approach for cancer prevention and treatment. Semin. Cancer Biol. 2015;35:S276–S304. doi: 10.1016/j.semcancer.2015.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Mallmann-Gottschalk N., Sax Y., Kimmig R., Lang S., Brandau S. EGFR-specific tyrosine kinase inhibitor modifies NK cell-mediated antitumoral activity against ovarian cancer cells. Int. J. Mol. Sci. 2019;20:4693. doi: 10.3390/ijms20194693. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Molecular Therapy Oncolytics are provided here courtesy of American Society of Gene & Cell Therapy

RESOURCES