The majority of ovarian cancer patients acquire resistance to standard platinum chemotherapy and novel therapies to reduce tumor burden and ascites accumulation are needed. Pregnancy-associated plasma protein-A (PAPP-A) plays a key role in promoting insulin-like growth factor (IGF) pathway activity, which directly correlates to ovarian cancer cell transformation, growth, and invasiveness. Herein, we evaluate PAPP-A expression in tumors and ascites of women with ovarian cancer, and determine the antitumor efficacy of a neutralizing monoclonal PAPP-A antibody (mAb-PA) in ovarian cancer using primary patient ovarian tumorgrafts (“Ovatars”). PAPP-A mRNA expression in patient ovarian tumors correlated with poor outcome and was validated as a prognostic surrogate in Ovatar tumors. Following confirmation of mAb-PA bioavailability and target efficacy in vivo, the antitumor efficacy of mAb-PA in multiple Ovatar tumor models was examined and the response was found to depend on PAPP-A expression. Strikingly, the addition of mAb-PA to standard platinum chemotherapy effectively sensitized platinum-resistant Ovatar tumors. PAPP-A protein in ascites was also assessed in a large cohort of patients and very high levels were evident across the entire sample set. Therefore, we evaluated targeted PAPP-A inhibition as a novel approach to managing ovarian ascites, and found that mAb-PA inhibited the development, attenuated the progression, and induced the regression of Ovatar ascites. Together, these data indicate PAPP-A as a potential palliative and adjunct therapeutic target for women with ovarian cancer. Mol Cancer Ther; 14(4); 973–81. ©2015 AACR.

Epithelial ovarian cancer is the leading cause of death among gynecologic malignancies (1). Although generally curable if detected at an early stage, the symptoms of ovarian cancer are vague and patients often present with advanced disease. The current standard for ovarian cancer includes primary surgical intervention to remove macroscopic disease burden (both solid tumor and any accompanying ascites) proceeded by a platinum-based chemotherapy regimen to abrogate any remaining microscopic deposits. Unfortunately, the majority of patients will relapse with platinum-resistant disease and develop intractable ascites, thereby contributing to the historically poor median overall survival (OS) rate. Thus, there is a critical need to identify novel targets and ways to predict response of the individual patient to new therapeutic agents to improve outcomes for women with this disease.

The role of insulin-like growth factors (IGF) in the development and progression of a broad range of epithelial cancers, including ovarian cancer, is well documented (2–4), and investigational compounds targeting the IGF axis have been developed for cancer therapeutics. In particular, monoclonal antibodies directed against the IGF-I receptor (IGF-IR), which transduces IGF-I and IGF-II signaling in cells, have been evaluated. Although conceptually sound, the clinical experience to date using these antibodies in unselected patients with various cancers has been largely disappointing. This can be explained by the broad-based activity of said antibodies (IGF-IR is ubiquitous and serves essential functions in normal tissues), lack of effect on IGF-II mitogenic signaling through insulin receptor isoform A (InsR-A), and secondary hormonal and metabolic derangements (3–7). Nevertheless, there remains the possible benefit to patient subgroups and, therefore, a need of predictive biomarkers to identify those patients who would be most likely to respond to specific IGF-targeted therapies. More importantly, alternative means of inhibiting IGF signaling to avoid the above side effects is desirable. We propose targeting pregnancy-associated plasma protein-A (PAPP-A).

PAPP-A is a novel metalloprotease of the metzincin superfamily (8). PAPP-A enhances IGF action through specific cleavage of inhibitory IGF binding proteins, primarily IGFBP-4, resulting in increased IGF bioavailability (reviewed in ref. 9). The secreted protease is tethered to the surface of the secreting and neighboring cells, thereby acting in an autocrine/paracrine manner to increase local IGF available for IGF-IR activation. It also has the potential to increase local IGF-II available for activation of InsR-A, an insulin receptor that is prevalent in tumor tissue and mediates a mitogenic signal (10). Conversely, therapeutic inhibition of PAPP-A proteolytic activity would effectively suppress both IGF-IR and InsR-A signaling, whereas insulin signaling through the classic InsR-B would not be affected by this approach. Thus, there are several advantages of targeting PAPP-A: It is a novel enzyme lending itself to specificity; it is present extracellularly, and therefore accessible; its expression is both condition- and cell-specific, and therefore selective; and its inhibition would suppress both IGF-I and IGF-II (but not insulin) signaling, thereby enhancing efficacy but limiting metabolic toxicity.

There are compelling data supporting PAPP-A inhibition of IGF signaling in ovarian cancer as a viable therapeutic approach. Several ovarian cancer cell lines and primary cultures express IGF-IR and have been shown to be responsive to IGFs (11, 12). In addition, IGF-II demonstrates mitogenic signaling through the InsR-A in human ovarian cancer cells (13). Wang and colleagues (14) found that an IGF-IR antibody inhibited ovarian cancer tumor growth in a xenograft model, and Gest and colleagues (15) suggest a role for IGF signaling in ovarian cancer aggressiveness. Moreover, we have recently shown that a relatively nontumorigenic ovarian cancer cell line becomes highly tumorigenic upon overexpression of PAPP-A (16). Conversely, decreased PAPP-A expression was associated with lowered invasive potential and tumor growth rate of an ovarian cancer cell line in vivo (17). Unfortunately, screening for PAPP-A expression in primary ovarian cancer has been limited (18, 19).

A substantial barrier to the study of ovarian cancer is the paucity of translationally and clinically relevant models. The development of primary patient ovarian tumorgrafts (“Ovatars”), with availability of source patient biospecimens (germline DNA, serum, frozen and formalin-fixed paraffin-embedded tissue) and prospective clinical annotations, helps to overcome these hurdles. We have shown that intraperitoneally derived Ovatars recapitulate patient tumor in terms of histologic, genomic, transcriptomic, and therapeutic heterogeneity (20). Thus, Ovatars represent a practical medium to study the effects of novel targets in ovarian cancer. Rather than selecting for clonal population of patient-derived cells able to grow in vitro, the generation of individualized orthotopic models allows for development and interaction of the tumor cells with the stroma in an environment similar to the source patient (20–22). As a result, experiments in Ovatars are more likely to produce clinically-relevant outcome parameters. To this end, we examined the potential role of PAPP-A as a prognostic surrogate of clinical outcome and predictive index of anti–PAPP-A–targeted therapy in patient ovarian cancer tumors and their respective Ovatars. Herein, we describe the efficacy of a novel PAPP-A–neutralizing antibody to limit tumor growth, prevent ascites accumulation and reverse platinum resistance in Ovatars.

Neutralizing PAPP-A monoclonal antibody

We have developed a high-affinity IgG monoclonal antibody against a substrate-binding exosite of PAPP-A required for proteolysis of IGFBP-4 (23). The development and characterization of this antibody and its effectiveness in inhibiting IGFBP-4 proteolysis and xenograft tumor growth has been published recently (24).

Ovatar model

The generation and expansion of viable ovarian tumor tissue obtained from consenting patients at the time of surgery has been described previously (20). Briefly, fresh patient tumor tissue was injected i.p. into severe combined immunodeficient (SCID) mice (Harlan). Upon engraftment, solid tumor (surgically resected and minced) or ascites was reimplanted into 20 to 80 mice, depending on the experiment, to generate biologic Ovatar replicates for in vivo experiments. The use of all human subject material was approved by the Institutional Review Board of Mayo Clinic. All animal studies were approved by the Institutional Animal Care and Use Committee of Mayo Clinic.

Treatments were initiated upon confirmation of tumors measuring ≥0.2 cm2 cross-sectional area or the presence of ascites as measured by transabdominal ultrasound (SonoSite S-series, SonoSite Inc.). Unless otherwise indicated, mice were treated weekly with monoclonal PAPP-A antibody (mAb-PA; 30 mg/kg) or IgG2a isotype control (Bio X Cell) via intraperitoneal delivery. For the platinum studies, Ovatars were randomized to receive i.p. saline or carboplatin plus paclitaxel (CP; NOVAPLUS) at 50 and 15 mg/kg, respectively, as described previously (20). Disease burden was assessed in tumor-bearing animals up to three times per week. After 4 weeks (or if clinical endpoints of tumor size, ascites burden, or morbidity were reached), mice were euthanized and blood and tumor tissue harvested. Final tumor weights were recorded and tumor sections snap frozen in liquid nitrogen. Where appropriate, ascites was collected, centrifuged, and acelluar and cellular components independently stored at −80°C. Personnel involved with acquisition of ultrasound measurements and subsequent tumor and/or ascites analyses were blinded to the treatments.

Microarray

For analysis of public microarray datasets, normalized gene-expression data were obtained from The Cancer Genome Atlas (TCGA) Research Network and Gene Expression Omnibus (GEO) database for the following independent studies: GSE13876, GSE14764, GSE49997, and GSE9891. Patient tumors within each cohort were ranked according to PAPPA expression and split evenly into two cohorts, defining the top 50% as “PAPPA high” and the bottom 50% as “PAPPA low.” Ovatar tumors (n = 118) were analyzed by Affymetrix HG U133 plus 2.0 arrays at the Mayo Medical Genome Facility according to the manufacturer's protocol. Gene-expression arrays were preprocessed and normalized by frozen multichip analysis (25). Patients were ranked according to their matched Ovatar PAPPA expression, split evenly into two groups (“PAPP-A high” vs. “PAPP-A low”) and assessed for outcome.

Immunohistochemistry

Formalin-fixed paraffin-embedded Ovatar tumor tissue samples were processed and immunostained for PAPP-A using a recombinant anti-human PAPP-A monoclonal antibody as previously described (26)

Human PAPP-A ELISA

Snap-frozen Ovatar tumor tissues were pulverized in liquid nitrogen using the Cellcrusher. PAPP-A protein levels in pulverized tumor tissue [lysed in M-PER extraction buffer (Thermo Scientific)] or acellular ascites were quantified using a highly sensitive PAPP-A ELISA (picoPAPP-A) generously provided by Ansh Laboratories. Of special note, this assay does not recognize mouse PAPP-A.

IGFBP-4 proteolysis

Aliquots of tumor lysates or cell-free ascites were incubated overnight at 37°C with IGFBP-4 and IGF-II (IGFBP-4 must bind IGF to be susceptible to cleavage by PAPP-A; ref. 27). IGFBP-4 proteolysis was assessed by Western blot analysis using primary antibodies toward the C- and N-termini of IGFBP-4 (Abcam) and fluorescently labeled secondary antibodies (LI-COR). Images were captured using the LI-COR Odyssey scanner and intensities quantitated using ImageJ software (28). IGFBP-4 proteolysis was also assessed using ELISA kits for total and intact IGFBP-4 (kindly provided by Ansh Laboratories). Total IGFBP-4 minus intact IGFBP-4 provides a quantitatively accurate measure of proteolyzed IGFBP-4.

IgG2 immunofluorescence

Cryosections (5 μm) of Ovatar tumor were fixed in methanol and dried to glass slides. Sections were rehydrated in PBS, blocked in protein-free buffer (Dako), and penetrated mAb-PA detected using a FITC-labeled anti-mouse IgG2.

IGF-I and IGFBP assessment

Total mouse IGF-I and total and active human IGF-I were measured using ELISA kits from Ansh Laboratories. The serum IGFBP profile was assessed by Western ligand blot analysis using radiolabeled IGF-I, as described previously (29).

Statistical analyses

Statistical significance between two groups was tested using ANOVA with the Bonferroni post hoc test for multiple comparison analysis using GraphPad Prism 6.0. Univariate analyses for progression-free survival (PFS) and OS were performed using the Kaplan–Meier method and corresponding log-rank test for intergroup differences. All analyses were conducted using JMP 9.0 (SAS Institute).

PAPP-A expression correlates with poor outcome in ovarian cancer tumors

To date, the prognostic value of PAPP-A expression in ovarian cancer tumors has not been reported. As an initial screen, five publicly available datasets were interrogated (TCGA, GSE13876, GSE14764, GSE49997, and GSE9891) for PAPPA gene expression split evenly into two cohorts, defining the top 50% as “PAPPA high” and the bottom 50% as “PAPPA low,” and correlated to patient outcome. Univariate HRs with corresponding 95% confidence intervals (CI) for PFS and OS were calculated and depicted in a forest plot (Fig. 1A). These data support high PAPPA as a direct correlate of poor outcome in terms of PFS and OS in ovarian cancer tumors. Moreover, univariate analysis of the combined cohorts yielded highly significant differences in PFS (HR, 1.581; 95% CI, 1.316–1.901; P < 0.001) and OS (HR, 1.558; 95% CI, 1.348–1.800; P < 0.0001; Fig. 1B).

Figure 1.

PAPPA expression correlates with poor outcome in patients with ovarian cancer and represents a prognostic surrogate in Ovatar tumors. A, the Forest plot of PFS (open) and OS (filled) by PAPPA expression in multiple ovarian carcinoma tumor cohorts, showing HRs (PAPPA low/high) and 95% CIs where an HR >1 implies a higher risk of recurrence and mortality in the PAPPA high group. Kaplan–Meier analyses of the combined cohorts depicting PFS and OS in the PAPPA low (gray line) versus PAPPA high (black line) for patient (B) and Ovatar (C) tumor expression.

Figure 1.

PAPPA expression correlates with poor outcome in patients with ovarian cancer and represents a prognostic surrogate in Ovatar tumors. A, the Forest plot of PFS (open) and OS (filled) by PAPPA expression in multiple ovarian carcinoma tumor cohorts, showing HRs (PAPPA low/high) and 95% CIs where an HR >1 implies a higher risk of recurrence and mortality in the PAPPA high group. Kaplan–Meier analyses of the combined cohorts depicting PFS and OS in the PAPPA low (gray line) versus PAPPA high (black line) for patient (B) and Ovatar (C) tumor expression.

Close modal

We recently demonstrated that Ovatar response to standard carboplatin/paxlitaxel chemotherapy recapitulates donor patient response (20). To further validate Ovatars as molecular surrogates, we sought to determine whether Ovatar PAPPA expression correlates to donor patient outcome. Patients were evenly stratified into two groups according to their Ovatar PAPPA expression as described above (PAPPA High vs. Low) and univariate analysis revealed significant differences in PFS (HR, 2.754; 95% CI, 1.147–6.615; P = 0.0235) and OS (3.433; 95% CI, 1.091–10.80; P = 0.0349; Fig. 1C).

Targeted PAPP-A inhibition confers antitumor efficacy in Ovatars

The following three Ovatars were selected from a cohort of 118 models, based on relative PAPPA expression, to test the effects of an inhibitory monoclonal antibody specific to PAPP-A (mAb-PA): PH006 (moderate expression, PH042 (low expression), and PH070 (high expression). Elevated PAPP-A expression was observed in 15% to 20% of ovarian tumors and was reflected in the Ovatars (Fig. 2A). In addition, PAPP-A protein levels were determined by ELISA (Table 1) and immunohistochemistry (Fig. 2B) and found to correlate with PAPPA gene expression.

Figure 2.

Antitumor efficacy of mAb-PA in Ovatars is dependent on PAPP-A levels. A, PAPPA expression in a cohort of Ovatar tumors (n = 118). Three Ovatar tumors (PH006, PH070, and PH042) were selected for in vivo testing (denoted by black bars). B, PAPP-A immunostaining of tumors from Ovatar PH006, PH070, and PH042. Tumor growth (normalized to pretreatment day 0; C) and corresponding Kaplan–Meier analysis (D) depicting OS in the mAb-PA–treated (gray line) versus IgG2a-treated (black line) PH006 (n = 7/group), PH070 (n = 10/group), and PH042 (n = 10/group) Ovatar models; error bars, SEM; asterisks denote statistical significance: *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 2.

Antitumor efficacy of mAb-PA in Ovatars is dependent on PAPP-A levels. A, PAPPA expression in a cohort of Ovatar tumors (n = 118). Three Ovatar tumors (PH006, PH070, and PH042) were selected for in vivo testing (denoted by black bars). B, PAPP-A immunostaining of tumors from Ovatar PH006, PH070, and PH042. Tumor growth (normalized to pretreatment day 0; C) and corresponding Kaplan–Meier analysis (D) depicting OS in the mAb-PA–treated (gray line) versus IgG2a-treated (black line) PH006 (n = 7/group), PH070 (n = 10/group), and PH042 (n = 10/group) Ovatar models; error bars, SEM; asterisks denote statistical significance: *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal
Table 1.

PAPP-A expression in Ovatar tumors and ascites

AvatarmRNAaTumorbAscitesc
PH006 Medium 0.7 d 
PH070 High 8.5 0.3 
PH042 Low nd nd 
AvatarmRNAaTumorbAscitesc
PH006 Medium 0.7 d 
PH070 High 8.5 0.3 
PH042 Low nd nd 

NOTE: PAPP-A protein levels in Ovatar tumors and ascites as measured by ultrasensitive PAPP-A ELISA.

Abbreviation: nd, not detectable.

aRelative mRNA expression (Fig. 2A).

bng/50 μg tumor.

cng/mL ascites.

dNo ascites available.

For the first experiment, Ovatar PH006 mice were treated with mAb-PA at varying doses (10 vs. 30 mg/kg, once vs. twice weekly) for a total of 4 weeks. At necropsy, all mice presented with multilobular tumors adherent to the pelvic floor. Final tumor weight was significantly reduced in mice receiving 30 mg/kg mAb-PA compared with IgG2a isotype control; there was no significant difference between the once versus twice weekly 30 mg/kg dose. In subsequent studies, significant inhibition of tumor growth (Fig. 2C) and increased OS (P = 0.0119; HR, 0.26; Fig. 2D) was demonstrated in Ovatar PH006 mice that received mAb-PA once weekly at 30 mg/kg compared with mice given IgG2a.

Treatment of Ovatar PH070 mice with mAb-PA once weekly at 30 mg/kg mAb-PA did not inhibit tumor growth (Fig. 2C), but significantly increased OS (P = 0.009; HR, 0.21; Fig. 2D). Investigation into the patient history of PH070 revealed that this Ovatar model recapitulated the clinical disease in terms of bowel adhesions and ascites development. PH070 Ovatars treated weekly with 30 mg/kg mAb-PA demonstrated decreased adhesions and ascites development (data not shown). Assessment of Ovatar tumors indicated penetrance of the mAb-PA (Fig. 3A) and effective inhibition of PAPP-A proteolytic activity toward IGFBP-4 (Fig. 3B).

Figure 3.

Bioavailability and target efficacy of mAb-PA in Ovatar tumors. A, immunofluorescent staining for the nucleus (blue) and IgG2a (green) in mAb-PA–treated (right) versus untreated (left) PH070 Ovatar tumors, indicating penetrance of the antibody into the tumor. B, Western blot analysis of intact (top, yellow) and proteolyzed (bottom, green/red) IGFBP-4 in IgG2a and mAb-PA–treated Ovatar tumors, indicating effective inhibition of PAPP-A activity in the tumor by mAb-PA.

Figure 3.

Bioavailability and target efficacy of mAb-PA in Ovatar tumors. A, immunofluorescent staining for the nucleus (blue) and IgG2a (green) in mAb-PA–treated (right) versus untreated (left) PH070 Ovatar tumors, indicating penetrance of the antibody into the tumor. B, Western blot analysis of intact (top, yellow) and proteolyzed (bottom, green/red) IGFBP-4 in IgG2a and mAb-PA–treated Ovatar tumors, indicating effective inhibition of PAPP-A activity in the tumor by mAb-PA.

Close modal

As a confirmatory measure of PAPP-A as a biomarker of therapeutic efficacy, we also evaluated the response of Ovatar PH042, which had relatively low PAPP-A mRNA expression and little or no PAPP-A protein (Table 1, Fig. 2B). mAb-PA did not significantly affect tumor progression (Fig. 2C) or outcome (Fig. 2D) in this Ovatar.

No apparent secondary effects of mAb-PA were observed

In all Ovatar models analyzed there were no significant differences in circulating levels of IGF-I (128 ± 7 ng/mL for mAb-PA vs.120 ± 10 ng/mL for IgG2a) or IGFBPs, the latter assessed by Western ligand blotting (data not shown).

mAb-PA treatment reduces ascites

To investigate the effect of mAb-PA on ascites accumulation and progression, it was determined that ultrasound was an accurate and precise measure of actual tumor weight and ascites volume (Supplementary Fig. S1). PH070 Ovatars were analyzed following tumor cell implantation. Ascites-free survival (AFS) was significantly increased (P < 0.0001; HR, 0.2351) and time for tumor-to-ascites appearance was significantly delayed (P < 0.0001) with mAb-PA treatment (Fig. 4A). To assess the effect of mAb-PA on ascites progression, a fraction of ascites containing active tumor cells from Ovatar PH070 was injected i.p. into mice, and mAb-PA or IgG2a treatments initiated 4 days later. OS was significantly increased (P = 0.0458; HR, 0.310) and ascites volume significantly decreased (P = 0.0317) in response to mAb-PA treatment (Fig. 4B). Moreover, mAb-PA treatment was found to induce ascites regression (P = 0.0157; Fig. 4C).

Figure 4.

Targeting PAPP-A in Ovatar models with ascites development. A, PAPP-A blockade inhibits ovarian ascites development. Left, Kaplan–Meier analysis depicting AFS in mAb-PA–treated (gray line) versus IgG2a-treated (black line) PH070 Ovatars (n = 40/group). Right, time (days) from solid tumor detection to the development of ascites in mAb-PA versus IgG2a-treated mice. B, PAPP-A blockade attenuates ovarian ascites progression. Left, Kaplan–Meier analysis depicting OS in PH070 Ovatars (n = 11/group) wherein treatment (mAb-PA vs. IgG2a) was initiated at the time of confirmed (ultrasound) subclinical ascites burden (day 0). Right, final ascites tumor burden depicted as volume in mL at the time of necropsy. C, PAPP-A blockade induces ovarian ascites regression. Ascites burden as measured by ultrasound (area in cm2) before treatment (Pre Tx) and after treatment (Post Tx) with mAb-PA versus IgG2a (n = 5/group).

Figure 4.

Targeting PAPP-A in Ovatar models with ascites development. A, PAPP-A blockade inhibits ovarian ascites development. Left, Kaplan–Meier analysis depicting AFS in mAb-PA–treated (gray line) versus IgG2a-treated (black line) PH070 Ovatars (n = 40/group). Right, time (days) from solid tumor detection to the development of ascites in mAb-PA versus IgG2a-treated mice. B, PAPP-A blockade attenuates ovarian ascites progression. Left, Kaplan–Meier analysis depicting OS in PH070 Ovatars (n = 11/group) wherein treatment (mAb-PA vs. IgG2a) was initiated at the time of confirmed (ultrasound) subclinical ascites burden (day 0). Right, final ascites tumor burden depicted as volume in mL at the time of necropsy. C, PAPP-A blockade induces ovarian ascites regression. Ascites burden as measured by ultrasound (area in cm2) before treatment (Pre Tx) and after treatment (Post Tx) with mAb-PA versus IgG2a (n = 5/group).

Close modal

PAPP-A levels in ovarian cancer patient ascites

The ascites collected from PH070 Ovatars had measureable levels of human PAPP-A, indicating production by the patient tumor. Although ascites from patient PH070 was not available, PAPP-A levels were evaluated in a large cohort of additional patient ascites. All patient ascites (N = 33) had high levels of PAPP-A, with mean ± SEM of 45 ± 3 ng/mL (normal serum levels < 1 ng/mL). Greater than 90% of the endogenous IGFBP-4 in the ascites was proteolyzed, and levels of bioactive IGF-I were 2% to 50% that of total IGF-I (mean ± SEM, 18 ± 3%). Proteolytic activity in patient ascites could be inhibited with mAb-PA (Fig. 5), demonstrating that PAPP-A is responsible for IGFBP-4 cleavage in this fluid.

Figure 5.

mAb-PA inhibits IGFBP-4 proteolysis in patient ascites. Aliquots of ascites from three different patients were incubated with IGFBP-4 and IgG2a or mAb-PA. Western blot analysis indicates effective inhibition of IGFBP-4 proteolysis by mAb-PA.

Figure 5.

mAb-PA inhibits IGFBP-4 proteolysis in patient ascites. Aliquots of ascites from three different patients were incubated with IGFBP-4 and IgG2a or mAb-PA. Western blot analysis indicates effective inhibition of IGFBP-4 proteolysis by mAb-PA.

Close modal

Combination mAb-PA and adjunct carboplatin/paclitaxel therapy

Patient PH070 had chemoresistant ovarian cancer (20). To determine whether targeted inhibition of PAPP-A could affect platinum resistance, standard platinum chemotherapy (CP) combined with mAb-PA therapy was tested (Fig. 6A). The combination CP plus mAb-PA versus CP plus IgG2a was initiated upon ultrasound confirmation of engrafted PH070 Ovatar tumors. Chemotherapy was ceased after 4 weeks and Ovatars continued to receive weekly treatments of mAb-PA or IgG2a for up to 100 days after initial treatment. As expected, CP treatment alone did not regress tumors below baseline during chemotherapy and the addition of IgG2a did not alter tumor growth trajectory. However, mAb-PA treatment enhanced sensitivity to platinum-based chemotherapy, as further supported by the highly significant reduction in final tumor weight (Fig. 6B).

Figure 6.

Adjuvant mAb-PA therapy sensitizes platinum-resistant Ovatars. A, tumor growth in response to platinum chemotherapy combined with mAb-PA [CP + mAb-PA, gray line] versus IgG2a [CP + IgG2a, black line] (n = 10/group). B, final tumor burden (solid tumor) at the time of necropsy.

Figure 6.

Adjuvant mAb-PA therapy sensitizes platinum-resistant Ovatars. A, tumor growth in response to platinum chemotherapy combined with mAb-PA [CP + mAb-PA, gray line] versus IgG2a [CP + IgG2a, black line] (n = 10/group). B, final tumor burden (solid tumor) at the time of necropsy.

Close modal

In this study, we demonstrate the efficacy of a novel neutralizing antibody against PAPP-A (mAb-PA) in primary patient ovarian tumorgrafts (Ovatars) to significantly: (i) inhibit intraperitoneal ovarian cancer tumor growth, (ii) delay ascites development, (iii) inhibit ascites accumulation, and (iv) induce ascites regression. Ovatars were selected for predictive response to treatment based on elevated PAPP-A expression in matched primary patient tumors. In contrast with the adverse effects reported in response to IGF-IR–directed antibodies (3–7), secondary endocrine disruption resulting from mAb-PA monotherapy was not evident. Furthermore, the addition of mAb-PA to standard frontline carboplatin/paclitaxel chemotherapy markedly improved tumor regression to effectively sensitize chemoresistant Ovatars.

Importance of the Ovatar model

Orthotopic implantation of patient ovarian cancer tissue in SCID mice results in the formation a primary tumor that is commonly localized to the mouse pelvis and/or ovaries. As these primary tumors develop and progress, cancer cell growth, proliferation, and metastatic dissemination within the peritoneal cavity is comparable with that of the patient. We have previously demonstrated that Ovatar tumors maintain the histopathologic and molecular diversity (genomic, transcriptomic, and proteomic) of the donor patient tumor (20). Moreover, Ovatars recapitulate the patient experience in terms of metastasis and ascites-related complications. Indeed, the Ovatar models (PH006, PH042, and PH070) used herein, exhibited markedly different characteristics across models while conserving the patient disease phenotype. For example, Patient PH006 presented with a solid stage III tumor with no ascites or signs of metastases at the time of primary debulking surgery whereas patient PH070 presented with adhesions, bowel involvement, and ascites. Thus, these orthotopic Ovatars more accurately resemble the patient experience in terms of clinical complications as compared with subcutaneous xenografts, and present a more relevant and directly translatable medium toward analyzing ovarian cancer disease progression and metastasis. More importantly, therapeutic response in Ovatars (standard and targeted therapeutics) should more likely reflect how the patient would respond, and, therefore, is a step closer toward translation into clinical benefit.

Necessity of identifying anti–PAPP-A therapy biomarkers

Arguably the greatest impedance to the advancement of IGF-IR–directed antibodies is a lack of predictive biomarkers, as the clinical value of IGF-IR remains controversial (2, 30). In this study, we used patient tumor PAPP-A gene expression to select candidate Ovatar models for testing mAb-PA therapy. DNA microarray data identified a subset of patient tumors (15%–20%) reporting high PAPP-A expression. Elevated PAPP-A protein expression in both solid tumors and in ascites was confirmed using a highly sensitive PAPP-A ELISA. Strikingly, 100% of patients' ascites tested reported high PAPP-A, and, with the exception of a single sample, levels were increased ≥100-fold compared with the average serum level of nonpregnant women. Bioactive IGF-I accounts for less than 1% of total IGF-I and high levels of free IGF-I are associated with disease progression (12). Virtually, all of the endogenous IGFBP-4 in patient ascites samples were found to be proteolyzed and, as a result, bioactive IGF-I levels were 2% to 50% of total IGF-I. Furthermore, we verified that PAPP-A present in the ascites remains proteolytically active. This was important as the expression of a naturally occurring, irreversible PAPP-A inhibitor (proMBP) has been shown to be increased in ovarian tumors and transformed ovarian epithelial cells (19).

Advantages of targeting PAPP-A via mAb-PA

The mAb-PA presented herein is uniquely specific to PAPP-A as it targets a substrate-binding exosite required for IGFBP-4 proteolysis (23). Importantly, the epitope of mAb-PA is not present in other enzymes, including homologous PAPP-A2 (primarily an IGFBP-5 protease; ref. 31). The specificity of this anti–PAPP-A therapy would, therefore, serve to improve the safety and tolerability via reducing off-target and potentially harmful side effects, and target inhibition to the site(s) of local (supraphyiologic) PAPP-A expression, in this case the tumor. This is in contrast with strategies targeting IGF-IR, IGF ligands, and IGFBPs that are ubiquitous, and therefore nonspecific to site or condition. Another advantage of mAb-PA is its potential to reduce both IGF-I and IGF-II binding to IGF-IR, IGF-IR:InsR hybrids, and InsR-A, while sparing any effect on insulin-stimulated InsR-B signaling. Therefore, there should be preferential reduction in proliferative/metastatic effects versus metabolic dysregulation.

Compensatory increases in circulating growth hormone (GH) and IGF-I have been observed with IGF-IR antibodies (4). There did not appear to be any secondary consequences of PAPP-A inhibition in the Ovatars resulting from mAb-PA. There were no apparent effects on either serum levels of IGF-I or profiling of IGFBPs, thus indirectly supporting that there were no effects on GH, which regulates serum IGF-I and IGFBP-3 (32). GH can also induce insulin resistance (33). We did not perform specific insulin sensitivity tests in Ovatars treated with mAb-PA, but previous studies in PAPP-A knockout mice did not indicate insulin resistance (34).

Ascites attenuation in response to mAb-PA treatment

Perhaps the most exciting finding in this study was the effect of mAb-PA therapy on inhibiting ascites development and accumulation as well as promoting regression of established ascites. Ascites produces significant morbidity in ovarian cancer, and palliative treatment options to reduce ascites burden are limited (35). Paracentesis via percutaneous drainage is frequently used for short-term symptom relief. Unfortunately, this invasive procedure carries many risks and frequently requires hospitalization. Thus, our findings suggest the potential use of mAb-PA monotherapy for palliative benefit, with minimal risk, toxicity, and discomfort.

In conjunction with previously cited work (16, 17), our Ovatar data implicate PAPP-A attenuation (expression/activity) as a potential strategy to limit and/or suppress ovarian cancer metastasis. Indeed, PAPP-A has recently been identified as a metastasis-related target gene (36, 37). It is of interest that ascites, which promotes tumor growth and metastases, has high levels of PAPP-A. Furthermore, ascites has been found to be rich in proinflammatory cytokines (38), which have been shown to upregulate PAPP-A expression in several cell types (39, 40). Further studies will be necessary to establish a role for PAPP-A in ovarian cancer metastases.

mAb-PA in platinum-resistant ovarian cancer

The application of potentially harmful and cytotoxic-targeted therapeutics in combination with standard chemotherapy in the frontline and/or maintenance setting has been rationalized by the promise of extending historically poor progression-free disease intervals and improving OS. For a subset of primary ovarian cancer tumors with elevated PAPP-A expression, anti–PAPP-A therapy presents as a viable adjuvant option in terms of favorable pharmacokinetic (e.g., easily administered, extensive tumor uptake, long half-life, little to no toxicity) and pharmacodynamic (e.g., low Kd, low IC50) attributes. Increased IGF signaling can potentially limit the efficacy of cytotoxic agents, and IGF-IR inhibition to overcome platinum resistance in ovarian cancer is by no means a novel concept (41, 42). PAPP-A is proposed as a better therapeutic target with greater tumor specificity and lower risk of side effects than other IGF system targets.

Identification of women with ovarian cancer who are most likely to respond is critical to the success of PAPP-A inhibition in clinical trials. In this study, we illustrate the potential power of patient selection through appropriate and easily obtainable biomarkers of PAPP-A in terms of expression and bioactivity, the efficacy of a novel neutralizing monoclonal antibody against PAPP-A, and proof-of-concept in a relevant patient Ovatar model supporting a multitude of therapeutics applications for PAPP-A as a promising new target in ovarian cancer.

M.A. Becker has ownership interest in Ovarian Cancer Tumorgrafts—Avatar System. P. Haluska has ownership interest in Mayo Ovarian Avatar-IP and is a consultant/advisory board member for Ansh. C. Oxvig has ownership interest in a patent application. C.A. Conover reports receiving a commercial research grant from Ansh Laboratories. No potential conflicts of interest were disclosed by the other author.

Conception and design: M.A. Becker, C. Oxvig, C.A. Conover

Development of methodology: M.A. Becker, P. Haluska Jr, C. Oxvig

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M.A. Becker, P. Haluska Jr, L.K. Bale

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M.A. Becker, P. Haluska Jr, C. Oxvig

Writing, review, and/or revision of the manuscript: M.A. Becker, P. Haluska Jr, C. Oxvig, C.A. Conover

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M.A. Becker, L.K. Bale

Study supervision: M.A. Becker, C.A. Conover

This work was supported by National Institutes of Health Endocrine Training grant T32 DK07352 (to M.A. Becker), Mayo Clinic Ovarian SPORE CA136393 (to M.A. Becker, P. Haluska, and C.A. Conover), and unrestricted research funding from Ansh Laboratories (to C.A. Conover).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Romero
I
,
Bast
RC
 Jr
. 
Minireview: human ovarian cancer: biology, current management, and paths to personalizing therapy
.
Endocrinology
2012
;
153
:
1593
602
.
2.
Samani
AA
,
Yakar
S
,
LeRoith
D
,
Brodt
P
. 
The role of the IGF system in cancer growth and metastasis: overview and recent insights
.
Endocr Rev
2007
;
28
:
20
47
.
3.
Weroha
SJ
,
Haluska
P
. 
IGF system in cancer
.
Endocrinol Metab Clin North Am
2012
;
41
:
335
50
.
4.
Pollak
M
. 
The insulin and insulin-like growth factor receptor family in neoplasia: an update
.
Nat Rev
2012
;
12
:
159
69
.
5.
King
H
,
Aleksic
A
,
Haluska
P
,
Macaulay
VM
. 
Can we unlock the potential of IGF-1R inhibition in cancer therapy?
Cancer Treat Rev
2014
;
40
:
1096
105
.
6.
Yee
D
. 
Insulin-like growth factor receptor inhibitors: baby or the bathwater?
J Natl Cancer Inst
2012
;
104
:
975
81
.
7.
Gao
J
,
Chang
YS
,
Jallal
B
,
Viner
J
. 
Targeting the insulin-like growth factor axis for the development of novel therapeutics in oncology
.
Cancer Res
2012
;
72
:
3
12
.
8.
Boldt
HB
,
Overgaard
MT
,
Laursen
LS
,
Weyer
K
,
Sottrup-Jensen
L
,
Oxvig
C
. 
Mutational analysis of the proteolytic domain of pregnancy-associated plasma protein-A (PAPP-A): classification as a metzincin
.
Biochem J
2001
;
358
:
359
67
.
9.
Conover
CA
. 
Key questions and answers about pregnancy-associated plasma protein-A
.
Trends Endocrinol Metab
2012
;
23
:
242
9
.
10.
Belfiore
A
,
Frasca
F
,
Pandini
G
,
Sciacca
L
,
Vigneri
R
. 
Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease
.
Endocr Rev
2009
;
30
:
586
623
.
11.
Kalli
KR
,
Conover
CA
. 
The insulin-like growth factor/insulin system in epithelial ovarian cancer
.
Front Biosci
2003
;
8
:
d714
22
.
12.
Brokaw
J
,
Katsaros
D
,
Wiley
A
,
Lu
L
,
Su
D
,
Sochirca
O
, et al
IGF-I in epithelial ovarian cancer and its role in disease progression
.
Growth Factors
2007
;
25
:
346
54
.
13.
Kalli
KR
,
Falowo
OI
,
Bale
LK
,
Zschunke
MA
,
Roche
PC
,
Conover
CA
. 
Functional insulin receptors on human epithelial ovarian carcinoma cells: implications for IGF-II mitogenic signaling
.
Endocrinology
2002
;
143
:
3259
67
.
14.
Wang
Y
,
Hailey
J
,
Williams
D
,
Wang
Y
,
Lipari
P
,
Malkowski
M
, et al
Inhibition of insulin-like growth factor-I receptor (IGF-IR) signaling and tumor cell growth by a fully human neutralizing anti-IGF-IR antibody
.
Mol Cancer Ther
2005
;
4
:
1214
21
.
15.
Gest
C
,
Mirshahi
P
,
Li
H
,
Pritchard
L-L
,
Joimel
U
,
Blot
E
, et al
Ovarian cancer: Stat3, RhoA and IGF-IR as therapeutic targets
.
Cancer Lett
2012
;
317
:
207
17
.
16.
Boldt
HB
,
Conover
CA
. 
Overexpression of pregnancy-associated plasma protein-A in ovarian cancer cells promotes tumor growth in vivo
.
Endocrinology
2011
;
152
:
1470
8
.
17.
Tanaka
Y
,
Kobayashi
H
,
Suzuki
M
,
Hirashima
Y
,
Kanayama
N
,
Terao
T
. 
Genetic downregulation of pregnancy-associated plasma protein-A (PAPP-A) by bikunin reduces IGF-I-dependent Akt and ERK1/2 activation and subsequently reduces ovarian cancer cell growth, invasion and metastasis
.
Int J Cancer
2004
;
109
:
336
47
.
18.
Alexiadis
M
,
Mamers
P
,
Chu
S
,
Fuller
PJ
. 
Insulin-like growth factor, insulin-like growth factor-binding protein-4, and pregnancy-associated plasma protein-A gene expression in human granulosa cell tumors
.
Int J Gynecol Cancer
2006
;
16
:
1973
9
.
19.
Kalli
KR
,
Chen
B-K
,
Bale
LK
,
Gernand
E
,
Overgaard
MT
,
Oxvig
C
, et al
Pregnancy-associated plasma protein-A (PAPP-A) expression and insulin-like growth factor binding protein-4 protease activity in normal and malignant ovarian surface epithelial cells
.
Int J Cancer
2004
;
110
:
633
40
.
20.
Weroha
SJ
,
Becker
MA
,
Enderica-Gonzalez
S
,
Harrington
S
,
Oberg
AL
,
Maurer
MJ
, et al
Tumorgrafts as in vivo surrogates for women with ovarian cancer
.
Clin Cancer Res
2014
;
20
:
1288
97
.
21.
Siolas
D
,
Hannon
GJ
. 
Patient-derived tumor xenografts: transforming clinical samples into mouse models
.
Cancer Res
2013
;
73
:
5315
9
.
22.
Mehner
C
,
Radisky
DC
. 
Triggering the landslide: the tumor-promotional effects of myofibroblasts
.
Exp Cell Res
2013
;
319
:
1657
62
.
23.
Mikkelsen
JH
,
Gyrup
C
,
Kristensen
P
,
Overgaard
MT
,
Poulsen
CB
,
Laursen
LS
, et al
Inhibition of the proteolytic activity of pregnancy-associated plasma protein-A by targeting substrate exosite binding
.
J Biol Chem
2008
;
283
:
16772
80
.
24.
Mikkelsen
JH
,
Resch
ZT
,
Kalra
B
,
Savjani
G
,
Kumar
A
,
Conover
CA
, et al
Indirect targeting of IGF receptor signaling in vivo by substrate-selective inhibition of PAPP-A
.
Oncotarget
2014
;
5
:
1014
25
.
25.
McCall
MN
,
Bolstad
BM
,
Irizarry
RA
. 
Frozen robust multiarray analysis (fRMA)
.
Biostatistics
2010
;
11
:
242
53
.
26.
Mansfield
AS
,
Visscher
DW
,
hart
SN
,
Wang
C
,
Goetz
MP
,
Oxvig
C
, et al
Pregnancy-associated plasma protein-A expression in human breast cancer
.
Growth Horm IGF Res
2014
;
24
:
264
7
.
27.
Qin
X
,
Byun
D
,
Lau
K-H
,
Baylink
DJ
,
Mohan
S
. 
Evidence that the interaction between insulin-like growth factor (IGF)-II and IGF binding protein (IGFBP)-4 is essential for the action of the IGF-II-dependent IGFBP-4 protease
.
Arch Biochem Biophys
2000
;
379
:
209
16
.
28.
Schneider
CA
,
Rasband
WS
,
Eliceiri
KW
. 
NIH Image to Image J: 25 years of image analysis
.
Nat Methods
2012
;
9
:
671
5
.
29.
Khosla
S
,
Hassoun
AAK
,
Baker
BK
,
Liu
F
,
Zein
NN
,
Whyte
MP
, et al
Insulin-like growth factor system abnormalities in hepatitis-associated osteosclerosis
.
J Clin Invest
1998
;
101
:
2165
73
.
30.
Werner
H
,
Maor
S
. 
The insulin-like growth factor-I receptor gene: a downstream target for oncogene and tumor suppressor action
.
Trends Endocrinol Metab
2006
;
17
:
236
42
.
31.
Overgaard
MT
,
Boldt
HB
,
Laursen
LS
,
Sottrup-Jensen
L
,
Conover
CA
,
Oxvig
C
. 
PAPP-A2, a novel insulin-like growth factor binding protein-5 proteinase
.
J Biol Chem
2001
;
276
:
21849
53
.
32.
Clemmons
DR
. 
Value of insulin-like growth factor system markers in the assessment of growth hormone status
.
Endocrinol Metab Clin
2007
;
36
:
109
29
.
33.
Dominici
FP
,
Argentino
DP
,
Munoz
MC
,
Miquet
JG
,
Sotelo
AI
,
Turyen
D
. 
Influence of the crosstalk between growth hormone and insulin signaling on the modulation of insulin sensitivity
.
Growth Horm IGF Res
2005
;
15
:
324
36
.
34.
Conover
CA
,
Mason
MA
,
Levine
JA
,
Novak
CM
. 
Metabolic consequences of pregnancy-associated plasma protein-A deficiency in mice: exploring possible relationship to the longevity phenotype
.
J Endocrinol
2008
;
198
:
599
605
.
35.
Kipps
E
,
Tan
DSP
,
Kaye
SB
. 
Meeting the challenge of ascites in ovarian cancer: new avenues for therapy and research
.
Nat Rev
2013
;
13
:
273
82
.
36.
Huang
J
,
Tabata
S
,
Kakiuchi
S
,
the Van
T
,
Goto
H
,
Hanibuchi
M
, et al
Identification of pregnancy-associated plasma protein A as a migration-promoting gene in malignant pleural mesothelioma cells: a potential therapeutic target
.
Oncotarget
2013
;
4
:
1172
84
.
37.
Salim
H
,
Arvanitis
A
,
de Petris
L
,
Kanter
L
,
Haag
P
,
Zovko
A
, et al
miRNA-214 is related to invasiveness of human non-small cell lung cancer and directly regulates alpha protein kinase 2 expression
.
Genes Chromosomes Cancer
2013
;
52
:
895
911
.
38.
Robinson Smith
TM
,
Isaacsohn
I
,
Mercer
CA
,
Zhou
M
,
Van Rooijen
N
,
Husseinzadeh
N
, et al
Macrophages mediate inflammation-enhanced metastasis of ovarian tumors in mice
.
Cancer Res
2007
;
67
:
5708
16
.
39.
Resch
ZT
,
Chen
B-K
,
Bale
LK
,
Oxvig
C
,
Overgaard
MT
,
Conover
CA
. 
Pregnancy-associated plasma protein A gene expression as a target of inflammatory cytokines
.
Endocrinology
2004
;
145
:
1124
9
.
40.
Boldt
HB
,
Conover
CA
. 
Pregnancy-associated plasma protein-A (PAPP-A): a local regulator of IGF bioavailability through cleavage of IGFBPs
.
Growth Horm IGF Res
2007
;
17
:
10
8
.
41.
Friedbichler
K
,
Hofmann
MH
,
Kroez
M
,
Ostermann
E
,
Lamche
HR
,
Koessl
C
, et al
Pharmacodynamic and antineoplastic activity of BI 836845, a fully human IGF ligand-neutralizing antibody, and mechanistic rationale for combination with rapamycin
.
Mol Cancer Ther
2014
;
13
:
399
409
.
42.
Eckstein
N
,
Servan
K
,
Hildebrandt
B
,
Politz
A
,
von Jonquieres
G
,
Wolf-Kummeth
S
, et al
Hyperactivation of the insulin-like growth factor receptor I signaling pathway is an essential event for cisplatin resistance of ovarian cancer cells
.
Cancer Res
2009
;
69
:
2996
3003
.

Supplementary data