VEGF pathway–targeting antiangiogenic drugs, such as bevacizumab, when combined with chemotherapy have changed clinical practice for the treatment of a broad spectrum of human cancers. However, adaptive resistance often develops, and one major mechanism is elevated tumor hypoxia and upregulated hypoxia-inducible factor-1α (HIF1α) caused by antiangiogenic treatment. Reduced tumor vessel numbers and function following antiangiogenic therapy may also affect intratumoral delivery of concurrently administered chemotherapy. Nonetheless, combining chemotherapy and bevacizumab can lead to improved response rates, progression-free survival, and sometimes, overall survival, the extent of which can partly depend on the chemotherapy backbone. A rational, complementing chemotherapy partner for combination with bevacizumab would not only reduce HIF1α to overcome hypoxia-induced resistance, but also improve tumor perfusion to maintain intratumoral drug delivery. Here, we evaluated bevacizumab and CRLX101, an investigational nanoparticle–drug conjugate containing camptothecin, in preclinical mouse models of orthotopic primary triple-negative breast tumor xenografts, including a patient-derived xenograft. We also evaluated long-term efficacy of CRLX101 and bevacizumab to treat postsurgical, advanced metastatic breast cancer in mice. CRLX101 alone and combined with bevacizumab was highly efficacious, leading to complete tumor regressions, reduced metastasis, and greatly extended survival of mice with metastatic disease. Moreover, CRLX101 led to improved tumor perfusion and reduced hypoxia, as measured by contrast-enhanced ultrasound and photoacoustic imaging. CRLX101 durably suppressed HIF1α, thus potentially counteracting undesirable effects of elevated tumor hypoxia caused by bevacizumab. Our preclinical results show pairing a potent cytotoxic nanoparticle chemotherapeutic that complements and improves concurrent antiangiogenic therapy may be a promising treatment strategy for metastatic breast cancer. Cancer Res; 76(15); 4493–503. ©2016 AACR.

Patients with triple-negative breast cancer (TNBC) have the highest risk of recurrence and metastasis (1). Various targeted therapies have been investigated, but as yet, none are currently approved. One notable example is bevacizumab, a VEGF-targeting antibody, which was granted accelerated FDA approval with weekly paclitaxel for first-line treatment of metastatic breast cancer, but this approval was later revoked after follow-up phase III clinical trials with different chemotherapy backbones showed less impressive benefits in progression-free survival (PFS; ref. 2). More recently, however, there has been renewed interest in reconsidering bevacizumab for the treatment of breast cancer based on several phase III clinical trial results, including one in the neoadjuvant and adjuvant setting (NSABP-B40) and two in the maintenance metastatic setting (IMELDA and TANIA; refs. 3–5). Importantly, a number of trial results and meta-analyses suggest the extent of beneficial effect (and associated toxicities) of adding bevacizumab to chemotherapy may depend on the concurrent chemotherapy regimen used (2, 4, 6, 7). Thus, an appropriate chemotherapy partner, one with better efficacy, manageable toxicity, and complementary modes of action, may be critical to gaining optimal benefit out of adding bevacizumab (and vice versa). One promising investigational chemotherapy drug is a nanoparticle–drug conjugate (NDC) known as CRLX101, which contains the payload camptothecin, a highly potent cytotoxic agent that inhibits topoisomerase-I (8, 9).

Camptothecin showed impressive preclinical antitumor activity but had low solubility, metabolic instability of the active lactone form, rapid renal clearance, and severe toxicities, which resulted in disappointing results in early-phase clinical trials (10, 11). A number of analogues were thus developed to improve the solubility of camptothecin, including the subsequently approved drugs, topotecan (Hycamtin, GlaxoSmithKline) and irinotecan (Camptosar, Pfizer; refs. 9, 12). Currently, there is no topoisomerase-I inhibitor approved for breast cancer treatment. Although both irinotecan and topotecan have been evaluated in phase II trials for metastatic breast cancer, they have not been approved as treatments for that indication given the associated high-grade toxicities experienced by patients for a modest benefit gain (13–17). Nonetheless, lessons learned from early trials using camptothecin (18), topotecan, and irinotecan suggest camptothecin analogues are indeed active in the treatment of breast cancer, but further clinical development requires better drug solubility, improved lactone ring stabilization, and less systemic toxicity. Notably, a recent phase III trial (BEACON) evaluated etirinotecan pegol, a new formulation of irinotecan, in patients with recurrent or metastatic breast cancer (19). It showed single-agent PFS benefits similar to treatment of physician's choice. Moreover, subgroup analyses showed etirinotecan pegol significantly prolonged overall survival in patients with a history of brain or liver metastases, and with two or more sites of disease (19). As an NDC of camptothecin, CRLX101 was designed to have superior solubility and stabilization of the lactone ring, as well as favorable safety and pharmacokinetics in patients (20, 21).

CRLX101 is composed of a cyclodextrin-containing polymer conjugated to camptothecin. In mice, the cyclodextrin polymer itself has no observable side effects or antitumor efficacy when tested up to 240 mg/kg (22). CRLX101 was designed to have improved accumulation within tumors by the enhanced permeability and retention (EPR) effect and thus reduced systemic exposure and toxicity (8, 20). The drug has been administered to more than 300 patients to date and appears to be generally well tolerated, achieving an overall response rate of 16% in 19 patients in a phase II clinical trial of platinum-resistant ovarian cancer as a monotherapy (23) and 23% in 22 patients in a phase I/II study of metastatic renal cell carcinoma in combination with bevacizumab (21).

We recently reported that CRLX101 plus bevacizumab resulted in synergistic antitumor efficacy in a preclinical model of advanced, metastatic ovarian cancer (23). CRLX101 was also shown to effectively and durably suppress elevated hypoxia-induced upregulation of hypoxia-inducible factor-1α (HIF1α) following therapy with bevacizumab, thus downregulating expression of downstream HIF1α-regulated markers, such as carbonic anhydrase IX (CAIX; ref. 23), and blocking the induction of cancer stem cells (24).

Here, we evaluated the combination of CRLX101 and bevacizumab in long-term therapy experiments of orthotopic primary TNBC xenografts either derived from tumor tissue fragment implantation of a patient-derived xenograft (PDX) called HCI-002 (25, 26) or cell injection of LM2-4, a luciferase-tagged variant of the established cell line MDA-MB-231 serially selected in vivo for aggressive spontaneous metastatic properties (27). We also evaluated long-term efficacy of CRLX101 and bevacizumab in a preclinical model of postsurgical, advanced metastatic TNBC (28), a model that recapitulates the more challenging clinical treatment of systemic metastatic disease (28, 29). We report that CRLX101 alone and in combination with bevacizumab is an effective treatment for advanced metastatic TNBC in these mouse models and provide new mechanistic results to help explain this encouraging antitumor activity.

Cell line and patient-derived tumor fragments

MDA-MB-231/LM2-4luc16+ (LM2-4) is a highly aggressive variant of MDA-MB-231 [parental line obtained from Jeff Lemontt (Genzyme Corp., Boston, MA) in 2000] and maintained in RPMI1640 supplemented with 5% FBS (27). LM2-4 was last authenticated in 2013 by Genetica DNA Laboratories (a LabCorp Specialty Testing Group) using analytical procedures for DNA extraction, PCR, and capillary electrophoresis on a 3130xl Genetic Analyzer (Applied Biosystems). The 13 core CODIS short tandem repeat (STR) loci plus PENTA E and PENTA D, and the gender-determining locus, amelogenin, were analyzed using the commercially available PowerPlex 16 HS Amplification Kit (Promega Corporation) and GeneMapper ID v3.2.1 software (Applied Biosystems) with appropriate positive and negative controls. Authentication of cell lines is confirmed by entering the STR DNA profile of each tested cell line into known repository cell line databases; authentication is defined as having a percent match with the reference STR profile ≥80% when using the ANSI/ATCC guidelines (ASN-0002-2011) or having a “unique” STR DNA profile (no matches found) for “in-house” cell lines not distributed by any cell line repository.

PDX HCI-002 tumor fragments were generously provided by Dr. Alana Welm (University of Utah, Salt Lake City, UT). Tumor fragments were originally obtained from a patient with TNBC and verified by histology to have retained morphology and characteristics as matched patient samples (25, 26). HCI-002 tumor fragments were serially passaged in SCID mice, and only early passages were used for all experiments.

Primary tumor implantation into the mammary fat pad

Eight-week-old female YFP-SCID mice were bred in-house. Procedures involving animals and their care were conducted in strict conformity with the guidelines of Sunnybrook Health Science Centre (Toronto, Ontario, Canada) and the Canadian Council of Animal Care. Mammary fat pad injections of LM2-4 (5 × 105 cells) and tumor fragment implantations (pieces of 2–5 mm3) of HCI-002 were carried out as described previously (26, 27). Mice were randomized by tumor volume prior to treatment initiation. All doses of CRLX101 are reported as camptothecin-equivalent doses.

Contrast-enhanced ultrasound photoacoustic imaging

In vivo imaging of primary tumors was conducted using a commercially available high-frequency laser-integrated ultrasound system (VevoLAZR, VisualSonics), which allowed for both photoacoustic and contrast-enhanced imaging (30). Contrast data were quantified with VevoCQ using time intensity curves (TIC) generated from the wash-in of microbubbles. Two parameters are taken from the TIC: peak enhancement and wash-in rate. Dual-wavelength PA imaging was used for real-time monitoring and calculation of oxygen saturation with the VevoLAB software.

Postsurgical, advanced metastatic breast cancer therapy model

LM2-4 cells were orthotopically implanted into the right mammary fat pad and resected when tumors reached 400 to 500 mm3, as described in ref. 27. Following primary tumor resection, distant visceral metastases can be detected by total body bioluminescence imaging using an IVIS200 Xenogen. Mice with metastatic disease were randomized on the basis of metastatic load and location before therapies were initiated (27, 29).

Statistical analysis

Results were reported as mean ± SD or SEM, as indicated. Tumor growth curves were reported as mean ± SD. Survival curves were plotted by the method of Kaplan and Meier and tested for survival differences with the log-rank test. Statistical significance was assessed by one-way ANOVA (Kruskal–Wallis test with Dunn post hoc) or t test (Mann–Whitney, two-tailed) using GraphPad Prism 4 (P < 0.05 was used as the threshold of statistical significance).

CRLX101 treatment can lead to complete primary tumor regressions

The MTD (8 mg/kg i.p. once weekly) of CRLX101 was previously established for SCID mice and is well tolerated even when combined with bevacizumab for long-term studies (23). In a primary tumor model derived from cell injection of LM2-4, CRLX101 caused rapid and durable tumor regressions (Fig. 1). Lower doses of CRLX101 monotherapy resulted in slower rates of tumor shrinkage compared with 8 mg/kg, resulting in small tumors that still eventually caused hind leg mobility impairment (smaller tumor volumes compared with vehicle but not statistically significant). Although the addition of bevacizumab to 2 mg/kg CRLX101 showed a trend for tumor growth delay, this was not statistically significant compared with 2 mg/kg CRLX101 monotherapy. Nonetheless, the addition of bevacizumab to 4 mg/kg CRLX101 significantly improved antitumor efficacy (P < 0.05 compared with 4 mg/kg CRLX101 alone), showing improved tumor growth suppression and shrinkage despite bevacizumab alone having no antitumor efficacy. However, despite early tumor suppression and continued therapy, bioluminescence imaging of mice treated with CRLX101 4 mg/kg plus bevacizumab showed these tumors eventually continued to grow. Notably, while mice were on-therapy continuously for 6 months, 8 mg/kg CRLX101 quickly and dramatically shrank established primary tumors, resulting in complete regressions (P < 0.001), with 3 of 5 mice essentially cured by 6 months of therapy (no observable bioluminescence signals). Similarly, 8 mg/kg CRLX101 plus bevacizumab also led to complete primary tumor regressions (P < 0.001 compared with vehicle, no statistically significant difference compared with 8 mg/kg CRLX101 monotherapy). Four of 5 mice treated with 8 mg/kg CRLX101 and bevacizumab did not have any residual disease or regrowth even after being off-therapy for an additional 4 months (Supplementary Fig. S1).

Figure 1.

CRLX101 caused regression of orthotopic primary breast tumors. A, SCID mice bearing LM2-4 tumors were treated with CRLX101 monotherapies (2, 4, and 8 mg/kg) and combined with bevacizumab (Bev). Therapy was started day 18 after tumor cell injection when tumors were approximately 200 to 250 mm3. Therapy was stopped after 6 months for mice treated with 8 mg/kg CRLX101 monotherapy or combination with bevacizumab. All other mice were treated until endpoint. Error bars, SD. n = 5 mice per group. B, all doses of CRLX101 significantly extended survival of mice, whereas bevacizumab monotherapy did not. ns, not significant.

Figure 1.

CRLX101 caused regression of orthotopic primary breast tumors. A, SCID mice bearing LM2-4 tumors were treated with CRLX101 monotherapies (2, 4, and 8 mg/kg) and combined with bevacizumab (Bev). Therapy was started day 18 after tumor cell injection when tumors were approximately 200 to 250 mm3. Therapy was stopped after 6 months for mice treated with 8 mg/kg CRLX101 monotherapy or combination with bevacizumab. All other mice were treated until endpoint. Error bars, SD. n = 5 mice per group. B, all doses of CRLX101 significantly extended survival of mice, whereas bevacizumab monotherapy did not. ns, not significant.

Close modal

Compared with primary tumors derived from LM2-4, PDX HCI-002 tumors were highly vascular. In contrast to bevacizumab's absence of efficacy when treating LM2-4 primary tumors, bevacizumab led to significantly delayed HCI-002 tumor growth while mice were on-therapy continuously for 5 months (P < 0.05 compared with vehicle; Fig. 2). Nonetheless, both doses of CRLX101 durably suppressed tumor growth (P = 0.002 for 4 mg/kg and P < 0.001 for 8 mg/kg CRLX101). Notably, the combination of either dose with bevacizumab significantly improved the antitumor efficacy compared with either drug alone (P < 0.001 compared with monotherapy). In this PDX model, despite tumor growth suppression or regression while mice were on-therapy, when all therapies were stopped after 5 months, tumor regrowth did occur but was delayed longer if mice were originally treated with a higher dose of CRLX101 or with concurrent bevacizumab.

Figure 2.

CRLX101 suppressed tumor growth of the PDX, HCI-002. A, SCID mice bearing orthotopic HCI-002 primary tumors were treated with 4 and 8 mg/kg CRLX101 as single agent or combined with bevacizumab (Bev). Therapy was started day 51 after implantation when tumors were approximately 200 to 250 mm3 and stopped after 5 months. Error bars, SD. n = 5 mice per group. B, all therapies, including bevacizumab monotherapy, significantly extended survival of mice.

Figure 2.

CRLX101 suppressed tumor growth of the PDX, HCI-002. A, SCID mice bearing orthotopic HCI-002 primary tumors were treated with 4 and 8 mg/kg CRLX101 as single agent or combined with bevacizumab (Bev). Therapy was started day 51 after implantation when tumors were approximately 200 to 250 mm3 and stopped after 5 months. Error bars, SD. n = 5 mice per group. B, all therapies, including bevacizumab monotherapy, significantly extended survival of mice.

Close modal

In separate experiments, tumors from mice treated for two weeks were evaluated for changes in microvessel density (MVD) and any corresponding changes in HIF1α (Fig. 3 and Supplementary Figs. S2 and S3). Bevacizumab reduced the number of tumor vessels by approximately 50% in LM2-4 tumors (Fig. 3A) and approximately 65% in HCI-002 tumors (Supplementary Fig. S2). This reduced MVD by bevacizumab monotherapy corresponded to an increase in HIF1α protein levels (Fig. 3B), which was confirmed by similar trends observed using immunohistochemical staining of HIF1α and CAIX, a downstream marker of HIF1α activity (Fig. 3C and D; ref. 23). In vitro, higher doses of CRLX101 showed a trend (although not statistically significant) of decreasing VEGF levels (Supplementary Fig. S3). However, in vivo, CRLX101 monotherapies did not significantly decrease MVD even though CRLX101 maintained low levels of HIF1α protein levels. Although CRLX101 monotherapies did not change the extent of tumor cell proliferation (Fig. 3E), there was a significant increase in apoptosis at all doses administered (Fig. 3F). Tumors from mice treated with concurrent bevacizumab and CRLX101 showed decreased MVD to levels similar to bevacizumab monotherapy. Nonetheless, concurrent CRLX101 was able to suppress any bevacizumab-induced increases in HIF1α. The reduced MVD observed for combination-treated tumors did not affect tumoral accumulation of CRLX101 (Supplementary Fig. S2). Interestingly, concurrent bevacizumab did not further increase the extent of apoptosis observed compared with CRLX101 monotherapies.

Figure 3.

CRLX101 therapy maintained low levels of HIF1α and significantly increased tumor cell apoptosis. After two weeks of therapy, tumors were stained for CD31 (A), processed for Western blot analysis to quantify HIF1α protein levels (n = 3 tumors/group; B), and stained for HIF1α (C) and CAIX (D). Bev, bevacizumab. Ki67 (E) and cleaved caspase-3 stainings (F) were used to assess changes in tumor cell proliferation and apoptosis, respectively. Error bars, SEM. *, P < 0.05; **, P < 0.01 compared with vehicle. n = 5 tumors per group for all stainings.

Figure 3.

CRLX101 therapy maintained low levels of HIF1α and significantly increased tumor cell apoptosis. After two weeks of therapy, tumors were stained for CD31 (A), processed for Western blot analysis to quantify HIF1α protein levels (n = 3 tumors/group; B), and stained for HIF1α (C) and CAIX (D). Bev, bevacizumab. Ki67 (E) and cleaved caspase-3 stainings (F) were used to assess changes in tumor cell proliferation and apoptosis, respectively. Error bars, SEM. *, P < 0.05; **, P < 0.01 compared with vehicle. n = 5 tumors per group for all stainings.

Close modal

We next evaluated whether the reduction in MVD by bevacizumab correlated with a change in functional tumor perfusion using in vivo contrast-enhanced ultrasound (CEUS) imaging, which uses gas-filled microbubbles to noninvasively monitor changes in blood flow and volume within tumors following therapy (Fig. 4; ref. 30). Tumors from mice treated with vehicle or bevacizumab alone showed decreased tumor blood volume and flow rate over the course of two weeks of therapy. In contrast, tumors from mice treated with CRLX101 monotherapy showed greatly improved perfusion, whereas perfusion in tumors treated with the combination remained unchanged. Parametric maps showing regions of high, low, or no perfusion confirm that tumors from mice treated with CRLX101 had smaller necrotic cores compared with vehicle and bevacizumab treatment. In the combination group, while a less perfused core was present, the surrounding “viable rim” remained well perfused. Similarly, CEUS imaging of PDX tumors showed CRLX101 maintained higher tumor perfusion and reduced tumor hypoxia, also confirmed by pimonidazole staining (Supplementary Fig. S4).

Figure 4.

CEUS and photoacoustic imaging of primary LM2-4 tumors. A, changes in blood flow volume (peak enhancement) and blood flow rate (wash-in rate) were measured using CEUS imaging. Bev, bevacizumab. Photoacoustic imaging was used to monitor changes in average tumor oxygen saturation. B, representative CEUS images of one tumor from each therapy group overlaid with parametric color mapping to show areas of high (red), low (blue), or no (black) perfusion. C, representative photoacoustic images of the same tumors overlaid with color mapping to indicate areas of high (red) and low (blue) tissue oxygenation. D, the number of vessels with open lumens was counted and reported as a percentage of the average number of vessels present per field. Vessels were counted in five randomly selected fields from five tumors per group (bracketed numbers, average number of vessels counted per field). Error bars, SEM. n = 4 to 6 tumors per group.*, P < 0.05; #, P = 0.07; **, P < 0.01 compared with vehicle.

Figure 4.

CEUS and photoacoustic imaging of primary LM2-4 tumors. A, changes in blood flow volume (peak enhancement) and blood flow rate (wash-in rate) were measured using CEUS imaging. Bev, bevacizumab. Photoacoustic imaging was used to monitor changes in average tumor oxygen saturation. B, representative CEUS images of one tumor from each therapy group overlaid with parametric color mapping to show areas of high (red), low (blue), or no (black) perfusion. C, representative photoacoustic images of the same tumors overlaid with color mapping to indicate areas of high (red) and low (blue) tissue oxygenation. D, the number of vessels with open lumens was counted and reported as a percentage of the average number of vessels present per field. Vessels were counted in five randomly selected fields from five tumors per group (bracketed numbers, average number of vessels counted per field). Error bars, SEM. n = 4 to 6 tumors per group.*, P < 0.05; #, P = 0.07; **, P < 0.01 compared with vehicle.

Close modal

In addition to measuring perfusion using CEUS, photoacoustic imaging was used to measure oxygen saturation (Fig. 4; ref. 30). Short laser pulses are directed at the tumor, generating thermoelastic expansion to create acoustic waves detected by an ultrasound transducer. Different absorption spectra of deoxygenated and oxygenated hemoglobin are then used to noninvasively estimate the spatial distribution of oxygen saturation in vivo (30). Consistent with CEUS data showing improved perfusion in CRLX101-treated tumors, these tumors had relatively higher tissue oxygenation levels compared with vehicle- and bevacizumab-treated tumors (Fig. 4C).

Improved perfusion with CRLX101 treatment suggests a more functional tumor vasculature, as results from Fig. 3A show CRLX101 does not result in an increase in the number of vessels. Tumor cells rapidly growing within a confined space causes compression (or “solid stress”) of tumoral blood vessels (31, 32). One possible explanation for the overall improved perfusion following CRLX101 is the drug causes extensive tumor cell apoptosis, which may then relieve compression of tumor blood vessels. To assess whether this may be the case, we evaluated whether CRLX101 resulted in more vessels with an open lumen, which would allow better blood flow within the tumor (Fig. 4D). Indeed, we observed that CRLX101 monotherapy resulted in a higher proportion of tumor blood vessels with open lumen compared with both vehicle and bevacizumab treatment. It is worth noting that in tumors treated with the drug combination, overall number of tumor vessels was reduced, but more of the remaining vessels had open lumens. These results were similarly confirmed in PDX tumors (Supplementary Fig. S5).

To obtain confirmation that CRLX101 relieves compression of tumor blood vessels by killing tumor cells, we evaluated whether tumor cell-packing density, that is, the number of cells found within a given area, changed following therapy (Fig. 5A). Although the average number of cells present within an imaged field was similar between vehicle- and bevacizumab-treated tumors, all doses of CRLX101 caused a significant reduction in cell-packing density. Fewer tumor cells densely packed within a tumor thus may result in fewer compressed tumor blood vessels and hence improved perfusion and oxygenation.

Figure 5.

CRLX101 reduced tumor cell density and resulted in fewer, smaller lung micrometastases. A, average number of nuclei counted within an imaged field (n = 5 tumors/group). Error bars, SEM. *, P < 0.05; **, P < 0.01 compared with vehicle. Bev, bevacizumab. B, lungs from mice still bearing primary tumors were collected after two weeks of therapy and stained for vimentin, used to specifically stain for human tumor cells. Lungs were classified as having no vimentin staining (and thus no tumor cells) present, only singly dispersed tumor cells, small clusters (<20 cells), or large clusters (>20 cells). For each therapy group, lungs from 5 mice were evaluated. Five serial sections were assessed for each lung, sectioned with 100 μm separation. Scale bar, 150 μm.

Figure 5.

CRLX101 reduced tumor cell density and resulted in fewer, smaller lung micrometastases. A, average number of nuclei counted within an imaged field (n = 5 tumors/group). Error bars, SEM. *, P < 0.05; **, P < 0.01 compared with vehicle. Bev, bevacizumab. B, lungs from mice still bearing primary tumors were collected after two weeks of therapy and stained for vimentin, used to specifically stain for human tumor cells. Lungs were classified as having no vimentin staining (and thus no tumor cells) present, only singly dispersed tumor cells, small clusters (<20 cells), or large clusters (>20 cells). For each therapy group, lungs from 5 mice were evaluated. Five serial sections were assessed for each lung, sectioned with 100 μm separation. Scale bar, 150 μm.

Close modal

Paradoxically, a more functional vasculature could result in a higher degree of tumor cell dissemination and metastasis to distant organs, such as the lungs. We therefore analyzed lungs from mice still bearing primary tumors for the presence of micrometastases or tumor cells that previously shed from the primary tumor and seeded in the lungs (Fig. 5B). Mice treated with bevacizumab monotherapy showed more micrometastases that were also larger in cluster size than those present in vehicle-treated mice. This is consistent with some previous preclinical reports that antiangiogenic agents used as monotherapies may cause an increase in metastasis and promote disease progression in mice despite an initial antitumor effect (33, 34). In stark contrast, mice treated with CRLX101 (particularly at higher doses) either showed no micrometastases or only singly dispersed tumor cells within the lungs, even when combined with bevacizumab. CRLX101 was therefore able to counteract potential prometastatic effects of bevacizumab. This effect is similar to published data where paclitaxel counteracted the proinvasive and prometastatic effects of DC101, the VEGFR-2 antibody (20), and metronomic topotecan reduced metastatic spread elicited by sunitinib (35).

CRLX101 in a postsurgical metastatic breast cancer model shrinks existing metastases and prevents the emergence of new metastases

Given the potency of CRLX101 in both primary tumor models and evidence showing CRLX101 was able to prevent formation of micrometastases, CRLX101 alone and with bevacizumab were evaluated in a postsurgical model of advanced metastatic breast cancer. Following surgical resection of established primary LM2-4 tumors, distant metastases were allowed to develop before treatment was initiated. Metastases were monitored by bioluminescence imaging and can manifest as lymph node metastases (causing mobility issues), lung metastases (leading to labored breathing), primary tumor regrowth or local metastases at the surgical site, as well as liver metastases and ascites (causing distended abdomens). Given the inherent variability of when and where metastases appear, every treatment group had randomized cohorts of mice with apparent distant metastases, local metastases or regrowths, as well as mice with no signs of metastases at the start of therapy (Fig. 6). Within 1.5 months of therapy, all mice treated with vehicle and bevacizumab monotherapy succumbed to disease, even if no apparent metastases were observed at the start of treatment. In contrast, bioluminescence imaging showed that in mice that had apparent metastases at the start of therapy, treatment with CRLX101 or CRLX101 plus bevacizumab caused these existing metastases to regress. In mice with no apparent metastases at the start of treatment, CRLX101 alone or combined with bevacizumab was able to prevent the emergence of new metastases both while mice were on-therapy (for 7 months) and then off-therapy (for 2 months). It should be noted, however, that if mice had a very heavy metastatic load in the lungs or ascites at the start of therapy, CRLX101 therapy was not as effective; we hypothesize this may be due to the need for some initial time for CRLX101 to accumulate within tumor cells and release its drug payload.

Figure 6.

CRLX101 prevented the emergence of new metastases and caused regression of existing metastases, thus greatly extending mice survival. A, bioluminescence images of 7 to 9 mice per group. All therapies were initiated 25 days after primary tumor resection and stopped after 7 months. B, all doses of CRLX101 significantly extended survival of mice, whereas bevacizumab monotherapy did not. ns, not significant.

Figure 6.

CRLX101 prevented the emergence of new metastases and caused regression of existing metastases, thus greatly extending mice survival. A, bioluminescence images of 7 to 9 mice per group. All therapies were initiated 25 days after primary tumor resection and stopped after 7 months. B, all doses of CRLX101 significantly extended survival of mice, whereas bevacizumab monotherapy did not. ns, not significant.

Close modal

CRLX101 thus significantly extended the survival of mice with metastatic disease (Fig. 6B). Although mice treated with vehicle or bevacizumab succumbed to metastases after 4 to 6 weeks, survival of mice treated with 4 mg/kg CRLX101 monotherapy was extended to 15 weeks. CRLX101 at 4 mg/kg plus bevacizumab was effective at durably suppressing metastasis. Notably, MTD CRLX101 was highly efficacious, so much so that any added benefit of combining it with bevacizumab was not detected. Upon necropsy, 3 of 9 mice (treated with 4 mg/kg CRLX101 plus bevacizumab), 5 of 8 mice (treated with 8 mg/kg CRLX101 alone), and 6 of 9 mice (treated with 8 mg/kg CRLX101 plus bevacizumab) were still alive and free of macroscopic metastases at the end of the experiment (7 months on-therapy and 2 months off-therapy).

We recently reported metronomic topotecan plus pazopanib delayed tumor growth of primary TNBC tumors and prolonged survival of mice with advanced metastatic disease (36). Given this promising therapy of combining a topoisomerase-I inhibitor (topotecan) and an antiangiogenic drug (pazopanib) for treating TNBC in mice, here, we evaluated CRLX101 and bevacizumab. There are several reasons why this alternative drug combination may be superior to that of topotecan and pazopanib. Single-agent topotecan had no antitumor efficacy and resulted in increased HIF1α. In contrast, single-agent CRLX101 is potently effective in not only delaying tumor growth, but causing marked and sustained tumor regressions. Furthermore, CRLX101 whether alone or combined with bevacizumab durably suppressed HIF1α. Finally, one important concern of combining topotecan and pazopanib is the tolerability and toxicity of both drugs, especially the combination, in the clinic (13, 14, 37). In contrast, current clinical data suggest that CRLX101 is tolerable in combination with standard doses of bevacizumab in patients (38, 39). The combination of CRLX101 and bevacizumab would thus appear to be more promising considering its lesser toxicity and better preclinical efficacy, an improved therapeutic index compared with the pazopanib/topotecan combination.

Limited efficacy successes of approved VEGF pathway–targeting antiangiogenic drugs may be due to several possible factors, including reduced intratumoral delivery of concurrently administered drugs, such as chemotherapy, as well as elevated hypoxia (and hence HIF1α), which contributes to resistance and may promote metastases (24, 40–42). Given these considerations, a complementary chemotherapy partner for combination with an antiangiogenic agent would be a highly potent agent that is able to improve tumor perfusion and reduce HIF1α without increasing tumor dissemination and/or metastases. Here, we showed that CRLX101 may be such a drug.

A number of strategies have been proposed, including vessel normalization (43) and “vascular promotion” therapy (31, 44), to improve intratumoral chemotherapy drug delivery. Alternatively, impaired perfusion, whether caused by antiangiogenic therapy or due to mechanical factors, such as tumor cells compressing vessels (31, 32), would be expected to reduce intratumoral delivery of chemotherapy drugs, such as CRLX101, in solid tumors (8, 45). However, CRLX101 maintained tumor perfusion despite concurrent antiangiogenic therapy. We report here that the potent antitumor activity of CRLX101 alleviated the solid stress by rapidly targeting tumor cells, in effect leading to major tumor regressions and decompressing tumor blood vessels to improve tumor perfusion [“tumor priming” (46)]. We and others have reported that metronomic dosing of a gemcitabine prodrug resulted in increased perfusion (47, 48). Paclitaxel-loaded tumor-penetrating microparticles have also been used to enhance siRNA delivery into solid tumors (46). Clinically, a similar effect was observed, where weekly paclitaxel reduced interstitial fluid pressure and improved tumor oxygenation (49). Interestingly, results from these studies and our work here with CRLX101 suggest chronic exposure of tumor cells to a cytotoxic drug, either by administering chemotherapy drugs in a frequent, metronomic fashion, or using drug formulations with intrinsically long half-lives, such as CRLX101, may be required to maintain sufficient durable tumor cell kill to persistently reduce compression of tumoral vessels and thus potentially improve perfusion. This improvement in tumor perfusion significantly reduces tumor hypoxia, which contributes to the ability of CRLX101 to effectively and durably suppress HIF1α. Even though HIF1α is elevated by tumor hypoxia, reduction in tumor hypoxia by improving tumor perfusion alone may not completely account for the HIF1α suppression by CRLX101. Rapisarda and colleagues have shown in vitro that metronomic topotecan was able to suppress HIF1α protein accumulation even when cells were maintained in normoxic conditions (50), suggesting HIF1α inhibition may occur even in the absence of elevated hypoxia. Taken together, we have shown that CRLX101 maintains or improves tumor perfusion and is able to durably suppress HIF1α protein levels even in the presence of bevacizumab, where tumors are more hypoxic, thus making CRLX101 an effective chemotherapy partner to pair with an antiangiogenic agent, such as bevacizumab.

One limitation of our results is the use of two primary tumor models and one metastatic model. Nevertheless, given our preclinical results of CRLX101 showing potent antitumor activity, particularly when treating advanced metastatic TNBC, and its complementary mode of action for combining with bevacizumab, CRLX101 plus bevacizumab may be a promising treatment strategy for breast cancer and other solid tumors.

C.G. Peters is a scientist at Cerulean Pharma. S. Eliasof has ownership interest (including patents) in Cerulean Pharma, Inc. F.S. Foster reports receiving a commercial research grant from and is a consultant/advisory board member for VisualSonics. R.S. Kerbel reports receiving other commercial research support from Cerulean and is a consultant/advisory board member for Cerulean and Triphase Accelerator. No potential conflicts of interest were disclosed by the other authors.

Conception and design: E. Pham, M. Yin, L. Jayaraman, R.S. Kerbel

Development of methodology: E. Pham, M. Yin, L. Jayaraman, E. Rohde, D. Lazarus, F.S. Foster

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): E. Pham, M. Yin, C.G. Peters, C.R. Lee, S. Man, E. Rohde, R.S. Kerbel

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): E. Pham, M. Yin, C.G. Peters, L. Jayaraman, R.S. Kerbel

Writing, review, and/or revision of the manuscript: E. Pham, M. Yin, C.G. Peters, P. Xu, L. Jayaraman, E. Rohde, D. Lazarus, S. Eliasof, F.S. Foster, R.S. Kerbel

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): E. Pham, C.R. Lee, S. Man, A. Chow

Study supervision: L. Jayaraman, S. Eliasof, F.S. Foster, R.S. Kerbel

Other (bioanalysis of study samples): D. Brown

The authors sincerely thank Dr. Marta Paez-Ribes for her assistance with animal surgeries and technical advice. The authors also thank Cassandra Cheng for outstanding administrative assistance and their student volunteer, Taylor VanVeen. Virtual slide scanning and image analysis consultation was provided by Taha Rashed at the Biomarker Imaging Research Laboratory (BIRL, Sunnybrook Research Institute) under the direction of Dr. Martin Yaffe. HCI-002 was generously provided by Dr. Alana Welm.

This work was supported by grants from the Canadian Institutes of Health Research (CIHR) and the Canadian Breast Cancer Foundation (R.S. Kerbel). Financial support was also provided by Cerulean Pharma Inc. R.S. Kerbel and F.S. Foster were recipients of Tier I Canada Research Chairs during the course of these studies. E. Pham is a recipient of a CIHR Post-Doctoral Fellowship Award.

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.
Foulkes
WD
,
Smith
IE
,
Reis-Filho
JS
. 
Triple-negative breast cancer
.
N Engl J Med
2010
;
363
:
1938
48
.
2.
Montero
AJ
,
Escobar
M
,
Lopes
G
,
Gluck
S
,
Vogel
C
. 
Bevacizumab in the treatment of metastatic breast cancer: friend or foe?
Curr Oncol Rep
2012
;
14
:
1
11
.
3.
Bear
HD
,
Tang
G
,
Rastogi
P
,
Geyer
CE
 Jr
,
Liu
Q
,
Robidoux
A
, et al
Neoadjuvant plus adjuvant bevacizumab in early breast cancer (NSABP B-40 [NRG Oncology]): secondary outcomes of a phase 3, randomised controlled trial
.
Lancet
2015
;
16
:
1037
48
.
4.
Gligorov
J
,
Doval
D
,
Bines
J
,
Alba
E
,
Cortes
P
,
Pierga
JY
, et al
Maintenance capecitabine and bevacizumab versus bevacizumab alone after initial first-line bevacizumab and docetaxel for patients with HER2-negative metastatic breast cancer (IMELDA): a randomised, open-label, phase 3 trial
.
Lancet
2014
;
15
:
1351
60
.
5.
von Minckwitz
G
,
Puglisi
F
,
Cortes
J
,
Vrdoljak
E
,
Marschner
N
,
Zielinski
C
, et al
Bevacizumab plus chemotherapy versus chemotherapy alone as second-line treatment for patients with HER2-negative locally recurrent or metastatic breast cancer after first-line treatment with bevacizumab plus chemotherapy (TANIA): an open-label, randomised phase 3 trial
.
Lancet
2014
;
15
:
1269
78
.
6.
Clark
O
,
Botrel
TE
,
Paladini
L
,
Ferreira
MB
. 
Targeted therapy in triple-negative metastatic breast cancer: a systematic review and meta-analysis
.
Core Evid
2014
;
9
:
1
11
.
7.
Fang
Y
,
Qu
X
,
Cheng
B
,
Chen
Y
,
Wang
Z
,
Chen
F
, et al
The efficacy and safety of bevacizumab combined with chemotherapy in treatment of HER2-negative metastatic breast cancer: a meta-analysis based on published phase III trials
.
Tumour Biol
2015
;
36
:
1933
41
.
8.
Eliasof
S
,
Lazarus
D
,
Peters
CG
,
Case
RI
,
Cole
RO
,
Hwang
J
, et al
Correlating preclinical animal studies and human clinical trials of a multifunctional, polymeric nanoparticle
.
Proc Natl Acad Sci U S A
2013
;
110
:
15127
32
.
9.
Svenson
S
,
Wolfgang
M
,
Hwang
J
,
Ryan
J
,
Eliasof
S
. 
Preclinical to clinical development of the novel camptothecin nanopharmaceutical CRLX101
.
J Control Release
2011
;
153
:
49
55
.
10.
Schaeppi
U
,
Fleischman
RW
,
Cooney
DA
. 
Toxicity of camptothecin (NSC-100880)
.
Cancer Chemother Rep
1974
;
5
:
25
36
.
11.
Beretta
GL
,
Gatti
L
,
Perego
P
,
Zaffaroni
N
. 
Camptothecin resistance in cancer: insights into the molecular mechanisms of a DNA-damaging drug
.
Curr Med Chem
2013
;
20
:
1541
65
.
12.
Zunino
F
,
Pratesi
G
. 
Camptothecins in clinical development
.
Expert Opin Investig Drugs
2004
;
13
:
269
84
.
13.
Kumler
I
,
Brunner
N
,
Stenvang
J
,
Balslev
E
,
Nielsen
DL
. 
A systematic review on topoisomerase 1 inhibition in the treatment of metastatic breast cancer
.
Breast Cancer Res Treat
2013
;
138
:
347
58
.
14.
Vanderbeeken
MC
,
Aftimos
PG
,
Awada
A
. 
Topoisomerase inhibitors in metastatic breast cancer: overview of current practice and future development
.
Curr Breast Cancer Rep
2013
;
5
:
31
41
.
15.
Wolff
AC
,
O'Neill
A
,
Kennedy
MJ
,
Stewart
JA
,
Gradishar
WJ
,
Lord
RS
 III
, et al
Single-agent topotecan as first-line chemotherapy in women with metastatic breast cancer: final results of eastern cooperative oncology group trial E8193
.
Clin Breast Cancer
2005
;
6
:
334
9
.
16.
Levine
EG
,
Cirrincione
CT
,
Szatrowski
TP
,
Canellos
G
,
Norton
L
,
Henderson
IC
. 
Phase II trial of topotecan in advanced breast cancer: a Cancer and Leukemia Group B study
.
Am J Clin Oncol
1999
;
22
:
218
22
.
17.
Perez
EA
,
Hillman
DW
,
Mailliard
JA
,
Ingle
JN
,
Ryan
JM
,
Fitch
TR
, et al
Randomized phase II study of two irinotecan schedules for patients with metastatic breast cancer refractory to an anthracycline, a taxane, or both
.
J Clin Oncol
2004
;
22
:
2849
55
.
18.
Natelson
EA
,
Giovanella
BC
,
Verschraegen
CF
,
Fehir
KM
,
De Ipolyi
PD
,
Harris
N
, et al
Phase I clinical and pharmacological studies of 20-(S)-camptothecin and 20-(S)-9-nitrocamptothecin as anticancer agents
.
Ann N Y Acad Sci
1996
;
803
:
224
30
.
19.
Perez
EA
,
Awada
A
,
O'Shaughnessy
J
,
Rugo
HS
,
Twelves
C
,
Im
SA
, et al
Etirinotecan pegol (NKTR-102) versus treatment of physician's choice in women with advanced breast cancer previously treated with an anthracycline, a taxane, and capecitabine (BEACON): a randomised, open-label, multicentre, phase 3 trial
.
Lancet
2015
;
16
:
1556
68
.
20.
Gaur
S
,
Wang
Y
,
Kretzner
L
,
Chen
L
,
Yen
T
,
Wu
X
, et al
Pharmacodynamic and pharmacogenomic study of the nanoparticle conjugate of camptothecin CRLX101 for the treatment of cancer
.
Nanomedicine
2014
;
10
:
1477
86
.
21.
Keefe
SM
,
Hoffman-Censits
JH
,
Mamtani
R
,
Walicki
M
,
Robinson
J
,
Smith
A
, et al
HIF inhibition in metastatic renal cell carcinoma (mRCC): final results of a phase Ib/IIa clinical trial evaluating the nanoparticle drug conjugate (NDC), CRLX101, in combination with bevacizumab (bev)
.
J Clin Oncol
33
, 
2015
(
suppl; abstr 4543
).
22.
Cheng
J
,
Khin
KT
,
Davis
ME
. 
Antitumor activity of beta-cyclodextrin polymer-camptothecin conjugates
.
Mol Pharm
2004
;
1
:
183
93
.
23.
Pham
E
,
Birrer
MJ
,
Eliasof
S
,
Garmey
EG
,
Lazarus
D
,
Lee
CR
, et al
Translational impact of nanoparticle-drug conjugate CRLX101 with or without bevacizumab in advanced ovarian cancer
.
Clin Cancer Res
2015
;
21
:
808
18
.
24.
Conley
SJ
,
Baker
TL
,
Burnett
JP
,
Theisen
RL
,
Lazarus
D
,
Peters
CG
, et al
CRLX101, an investigational camptothecin-containing nanoparticle-drug conjugate, targets cancer stem cells and impedes resistance to antiangiogenic therapy in mouse models of breast cancer
.
Breast Cancer Res Treat
2015
;
150
:
559
67
.
25.
Paez-Ribes
M
,
Man
S
,
Xu
P
,
Kerbel
RS
. 
Potential pro-invasive or metastatic effects of preclinical antiangiogenic therapy are prevented by concurrent chemotherapy
.
Clin Cancer Res
2015
;
21
:
5488
98
.
26.
DeRose
YS
,
Wang
G
,
Lin
YC
,
Bernard
PS
,
Buys
SS
,
Ebbert
MT
, et al
Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes
.
Nat Med
2011
;
17
:
1514
20
.
27.
Munoz
R
,
Man
S
,
Shaked
Y
,
Lee
CR
,
Wong
J
,
Francia
G
, et al
Highly efficacious nontoxic preclinical treatment for advanced metastatic breast cancer using combination oral UFT-cyclophosphamide metronomic chemotherapy
.
Cancer Res
2006
;
66
:
3386
91
.
28.
Man
S
,
Bocci
G
,
Francia
G
,
Green
SK
,
Jothy
S
,
Hanahan
D
, et al
Antitumor effects in mice of low-dose (metronomic) cyclophosphamide administered continuously through the drinking water
.
Cancer Res
2002
;
62
:
2731
5
.
29.
Guerin
E
,
Man
S
,
Xu
P
,
Kerbel
RS
. 
A model of postsurgical advanced metastatic breast cancer more accurately replicates the clinical efficacy of antiangiogenic drugs
.
Cancer Res
2013
;
73
:
2743
8
.
30.
Yin
M
,
Lakshman
M
,
Foster
FS
. 
Quantitative functional assessment of tumour microenvironment using contrast enhanced ultrasound and photoacoustic imaging [abstract]
. In:
Proceedings of the 2013 IEEE International Ultrasonics Symposium (IUS); 2013 Jul 21-25; Prague, Czech Republic
.
New York (NY)
:
IEEE
; 
2013
.
p.
1833
36
.
31.
Stylianopoulos
T
,
Jain
RK
. 
Combining two strategies to improve perfusion and drug delivery in solid tumors
.
Proc Natl Acad Sci U S A
2013
;
110
:
18632
7
.
32.
Stylianopoulos
T
,
Martin
JD
,
Chauhan
VP
,
Jain
SR
,
Diop-Frimpong
B
,
Bardeesy
N
, et al
Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors
.
Proc Natl Acad Sci U S A
2012
;
109
:
15101
8
.
33.
Paez-Ribes
M
,
Allen
E
,
Hudock
J
,
Takeda
T
,
Okuyama
H
,
Vinals
F
, et al
Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis
.
Cancer Cell
2009
;
15
:
220
31
.
34.
Ebos
JM
,
Lee
CR
,
Cruz-Munoz
W
,
Bjarnason
GA
,
Christensen
JG
,
Kerbel
RS
. 
Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis
.
Cancer Cell
2009
;
15
:
232
9
.
35.
Rovida
A
,
Castiglioni
V
,
Decio
A
,
Scarlato
V
,
Scanziani
E
,
Giavazzi
R
, et al
Chemotherapy counteracts metastatic dissemination induced by antiangiogenic treatment in mice
.
Mol Cancer Ther
2013
;
12
:
2237
47
.
36.
Di Desidero
T
,
Xu
P
,
Man
S
,
Bocci
G
,
Kerbel
RS
. 
Potent efficacy of metronomic topotecan and pazopanib combination therapy in preclinical models of primary or late stage metastatic triple-negative breast cancer
.
Oncotarget
2015
;
6
:
42396
410
.
37.
Burris
HA
 III
,
Dowlati
A
,
Moss
RA
,
Infante
JR
,
Jones
SF
,
Spigel
DR
, et al
Phase I study of pazopanib in combination with paclitaxel and carboplatin given every 21 days in patients with advanced solid tumors
.
Mol Cancer Ther
2012
;
11
:
1820
8
.
38.
Keefe
SM
,
Hennessey
M
,
Gunnarsson
O
,
Mamtani
R
,
Vaughn
DJ
,
Hoffman-Censits
JH
, et al
Phase 1b/2a study of the nanopharmaceutical CRLX101 with bevacizumab (bev) in the treatment of patients (pts) with refractory metastatic renal cell carcinoma (mRCC): results from the planned interim analysis [abstract]
. In:
Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA
.
AACR
:
Philadelphia (PA)
; 
2014
.
Abstract nr CT340
.
39.
Weiss
GJ
,
Chao
J
,
Neidhart
JD
,
Ramanathan
RK
,
Bassett
D
,
Neidhart
JA
, et al
First-in-human phase 1/2a trial of CRLX101, a cyclodextrin-containing polymer-camptothecin nanopharmaceutical in patients with advanced solid tumor malignancies
.
Invest New Drugs
2013
;
31
:
986
1000
.
40.
Rapisarda
A
,
Melillo
G
. 
Overcoming disappointing results with antiangiogenic therapy by targeting hypoxia
.
Nat Rev Clin Oncol
2012
;
9
:
378
90
.
41.
Semenza
GL
. 
Cancer-stromal cell interactions mediated by hypoxia-inducible factors promote angiogenesis, lymphangiogenesis, and metastasis
.
Oncogene
2013
;
32
:
4057
63
.
42.
Van der Veldt
AA
,
Lubberink
M
,
Bahce
I
,
Walraven
M
,
de Boer
MP
,
Greuter
HN
, et al
Rapid decrease in delivery of chemotherapy to tumors after anti-VEGF therapy: implications for scheduling of anti-angiogenic drugs
.
Cancer Cell
2012
;
21
:
82
91
.
43.
Tredan
O
,
Lacroix-Triki
M
,
Guiu
S
,
Mouret-Reynier
MA
,
Barriere
J
,
Bidard
FC
, et al
Angiogenesis and tumor microenvironment: bevacizumab in the breast cancer model
.
Target Oncol
2015
;
10
:
189
98
.
44.
Wong
PP
,
Demircioglu
F
,
Ghazaly
E
,
Alrawashdeh
W
,
Stratford
MR
,
Scudamore
CL
, et al
Dual-action combination therapy enhances angiogenesis while reducing tumor growth and spread
.
Cancer Cell
2015
;
27
:
123
37
.
45.
Maeda
H
,
Nakamura
H
,
Fang
J
. 
The EPR effect for macromolecular drug delivery to solid tumors: improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo
.
Adv Drug Deliv Rev
2013
;
65
:
71
9
.
46.
Wang
J
,
Lu
Z
,
Yeung
BZ
,
Wientjes
MG
,
Cole
DJ
,
Au
JL
. 
Tumor priming enhances siRNA delivery and transfection in intraperitoneal tumors
.
J Control Release
2014
;
178
:
79
85
.
47.
Francia
G
,
Shaked
Y
,
Hashimoto
K
,
Sun
J
,
Yin
M
,
Cesta
C
, et al
Low-dose metronomic oral dosing of a prodrug of gemcitabine (LY2334737) causes antitumor effects in the absence of inhibition of systemic vasculogenesis
.
Mol Cancer Ther
2012
;
11
:
680
9
.
48.
Cham
KK
,
Baker
JH
,
Takhar
KS
,
Flexman
JA
,
Wong
MQ
,
Owen
DA
, et al
Metronomic gemcitabine suppresses tumour growth, improves perfusion, and reduces hypoxia in human pancreatic ductal adenocarcinoma
.
Br J Cancer
2010
;
103
:
52
60
.
49.
Taghian
AG
,
Abi-Raad
R
,
Assaad
SI
,
Casty
A
,
Ancukiewicz
M
,
Yeh
E
, et al
Paclitaxel decreases the interstitial fluid pressure and improves oxygenation in breast cancers in patients treated with neoadjuvant chemotherapy: clinical implications
.
J Clin Oncol
2005
;
23
:
1951
61
.
50.
Rapisarda
A
,
Uranchimeg
B
,
Sordet
O
,
Pommier
Y
,
Shoemaker
RH
,
Melillo
G
. 
Topoisomerase I-mediated inhibition of hypoxia-inducible factor 1: mechanism and therapeutic implications
.
Cancer Res
2004
;
64
:
1475
82
.