Cancer Treatment Reviews 40 (2014) 578–584
Contents lists available at ScienceDirect
Cancer Treatment Reviews journal homepage: www.elsevierhealth.com/journals/ctrv
New Drugs
Liposomal nanomedicines in the treatment of prostate cancer Jan Kroon a,b,1, Josbert M. Metselaar b,2, Gert Storm b,c,3,4, Gabri van der Pluijm a,⇑ a
Department of Urology, Leiden University Medical Center, Leiden, The Netherlands Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands c Department of Pharmaceutics, Utrecht University, Utrecht, The Netherlands b
a r t i c l e
i n f o
Article history: Received 23 August 2013 Received in revised form 10 October 2013 Accepted 16 October 2013
Keywords: Active targeting Liposomes Passive targeting Prostate cancer Nanomedicine
a b s t r a c t Prostate cancer is the most common cancer type and the second leading cause of death from cancer in males. In most cases, no curative treatment options are available for metastatic castration-resistant prostate cancer as these tumors are highly resistant to chemotherapy. Targeted drug delivery, using liposomal drug delivery systems, is an attractive approach to enhance the efficacy of anticancer drugs and prevent side effects, thereby potentially increasing the therapeutic index. In most preclinical prostate cancer studies, passive liposomal targeting of anticancer drugs (caused by enhanced permeability and retention of the therapeutic compound) leads to an increased antitumor efficacy and decreased side effects compared to non-targeted drugs. As a result, the total effective dose of anticancer drugs can be substantially decreased. Active (ligand-mediated) liposomal targeting of tumor cells and/or tumor-associated stromal cells display beneficial effects, but only limited preclinical studies were reported. To date, clinical studies in prostate carcinoma have been performed with liposomal doxorubicin only. These studies showed that long-circulating, PEGylated, liposomal doxorubicin generally outperforms conventional short-circulating liposomal doxorubicin, stressing the importance of passive tumor targeting for this drug in prostate carcinoma. In this review, we provide an overview of the (pre)clinical studies that focus on liposomal drug delivery in prostate carcinoma. Ó 2013 Elsevier Ltd. All rights reserved.
Introduction on prostate cancer and liposomal drug delivery Over the past decades, substantial progress has been made in the field of nanomedicinal drug delivery [1,2]. In this booming field, liposomes have taken a front-runner position and have been evaluated extensively in preclinical and clinical cancer settings. Meanwhile, a few liposomal formulations have been clinically approved for the treatment of cancer [3]. Among the extensive amount of studies in the field of liposomal tumor targeting, only a limited number of investigations have focused on the utility of ⇑ Corresponding author. Address: Head of Uro-Oncology Research Laboratory, J3100, Department of Urology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands. Tel.: +31 71 5265275. E-mail addresses:
[email protected] (J. Kroon),
[email protected] (J.M. Metselaar),
[email protected] (G. Storm),
[email protected] (G. van der Pluijm). 1 Address: J3-64, Department of Urology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands. Tel.: +31 71 5266140. 2 Address: Department of Targeted Therapeutics, MIRA Institute for Biological Technology and Technical Medicine, Drienernolaan 5, 7522 NB, Enschede, The Netherlands. Tel.: +31 53 4893096. 3 Address: Department of Targeted Therapeutics, MIRA Institute for Biological Technology and Technical Medicine, Drienernolaan 5, 7522 NB, Enschede, The Netherlands. Tel.: +31 53 4892832. 4 Address: Department of Pharmaceutical Sciences, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands. Tel.: +31 30 2537388. 0305-7372/$ - see front matter Ó 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.ctrv.2013.10.005
liposomes in prostate cancer treatment. It is striking that amongst those studies, castration-resistant prostate cancer (CRPC) has deserved relatively little attention, as CRPC is one of the most detrimental among the advanced-stage cancers, with very little effective treatment options currently available. Many drugs designed for the treatment of CRPC fail at some point during clinical development due to intrinsic/acquired resistance and/or dose-limiting side effects. Described mechanisms for therapy resistance include overexpression of P-glycoprotein [4] and enhanced STAT1 expression [5]. Targeted drug delivery systems like liposomes may help overcome drug resistance as higher drug levels are potentially achievable at the tumor site. In addition, targeted drug delivery can diminish drug exposure of healthy tissues leading to less systemic side effects. In light of the extensive experience with several liposomal anticancer formulations [6], liposomal targeting of anticancer drugs to tumors in patients with prostate cancer seems a plausible drug targeting approach. Liposomes are versatile, self-assembling, carrier materials that contain one or more lipid bilayers with phospholipids and/or cholesterol as major lipid components, and can be used to encapsulate hydrophilic drugs in their inner aqueous compartment(s) while more hydrophobic drugs can associate with the lipid bilayer(s) (Fig. 1) (reviewed in [7,8]). Compared to other nanocarriers, liposomes are relatively easy to prepare, biodegradable and essentially
J. Kroon et al. / Cancer Treatment Reviews 40 (2014) 578–584
Fig. 1. Structure of a liposome used as drug delivery system.
nontoxic, although size is usually limited to 50–150 nm if used for drug delivery purposes [9,10]. Liposomes have been shown useful for drugs with unfavorable pharmacokinetic properties that result in a suboptimal therapeutic index. The addition of a polyethylene glycol (PEG) coating to the outer surface has been a major breakthrough as this coating opposes detection by the mononuclear phagocyte system (MPS) and thereby strongly enhances circulation time of intravenously injected liposome particles. As tumors often display a chaotic and highly permeable vasculature as a result of angiogenesis, the long circulation time of PEG-liposomes allows enhanced extravasation of liposomes into the tumor microenvironment compared to healthy tissues. Generally, an increased liposomal size favors extravasation as long as this size does not exceed the size of the inter-endothelial fenestrae, which are typically 200–400 nm [11–13]. After extravasation, liposomes are usually retained since lymphatic drainage is often impaired in tumors [7]. Hence, this tumor targeting mechanism is referred to as the enhanced permeability and retention (EPR) effect. Because no specific targeting ligands are used to interact with the tumor target site, this tumor localization process is referred to as ’passive targeting’ and represents the major targeting principle for intravenously administered long-circulating liposomes (Fig. 2, upper left) [7,8]. Conversely, ’active targeting’ implies a ligand or antibody bound to the outer surface of liposomes that selectively target receptors/ligands overexpressed on the tumor cells (Fig. 2, upper right) or the (a)cellular tumor microenvironment (Fig. 2, lower left) [7,8,14]. Following binding to the receptor, internalization via receptor-mediated endocytosis can take place. Both the extent of tumor localization and subsequent cellular internalization determine the therapeutic efficacy of liposome-encapsulated anticancer agents [15]. The aim of this review is to summarize the literature on both passively and actively targeted liposomes for the treatment of prostate cancer, and to provide a perspective on the use of targeted liposomes as a new therapeutic option to treat this malignancy. Preclinical studies A limited number of studies focused on passive and/or active liposomal targeting of chemotherapeutic agents in preclinical
579
prostate cancer models. Chemotherapy is widely used to treat prostate carcinoma, but is reserved only for the later stages of the disease, when the disease has progressed into the stage of CRPC for which typically a combination of docetaxel and prednisone is given [16,17]. Unfortunately, only a small proportion of patients respond to docetaxel and dose-limiting myelosuppression prohibits intensification of treatment [16]. This unfavorable situation provides a strong rationale for tumor-targeted delivery of chemotherapeutic agents. A phase I study with liposomal docetaxel was conducted in a cohort of multiple advanced solid malignancies which revealed higher maximum tolerated dosages of the liposomal formulation compared to free docetaxel (85 mg/m2, or 110 mg/m2 with G-SCF support; compared to 75 mg/m2 for free docetaxel) [18]. Surprisingly, while being the standard-of-care for CRPC, liposomal docetaxel has not yet been investigated in preclinical models of prostate cancer. This is even more striking considering the range of studies that have been performed with liposomal formulations of other chemotherapeutic agents, including doxorubicin [19–23], gemcitabine [24,25], paclitaxel [26] and mitoxantrone [27]. Doxorubicin, an anthracycline widely used as chemotherapeutic agent, is associated with several side effects, most notably cardiotoxicity [28], and liposomal delivery of doxorubicin was proven useful to reduce chronic cardiotoxicity. As a result, liposomal delivery increases the therapeutic index of the drug. Indeed, liposomal doxorubicin has been clinically approved for the treatment of Kaposi’s sarcoma, ovarian cancer, breast cancer and multiple myeloma (as PEG-liposomal doxorubicin marketed as Doxil in the USA and Caelyx outside the USA) and for advanced breast cancer (the non-PEGylated liposomal doxorubicin version marketed as Myocet) [3,29]. Passive delivery of liposomal doxorubicin was examined in multiple human prostate cancer cell line-based and primary prostate cancer-based in vivo models. Monotherapy with liposomal doxorubicin resulted in contrasting results, with three studies showing significant inhibition of subcutaneous tumor growth [19–21] while one study showed no effect [22]. It is hard to pinpoint the reason for these differential responses, as there were differences in liposomal compositions, size, tumor models, dosing and time of treatment. Liposomal delivery of gemcitabine, a nucleoside analog clinically used for several types of cancer, induced a potent antitumor effect which could only be matched by 45-fold higher doses of free gemcitabine (8 mg/kg/week versus 360 mg/kg/week, respectively) [24,25]. Moreover, decreased numbers of lymph node metastases were observed upon treatment with liposomal gemcitabine compared to free gemcitabine [25]. Liposomal delivery of mitoxantrone, the previous second-line treatment for CRPC, showed an inhibition of prostate xenograft growth but was not compared to free mitoxantrone [27]. In contrast to doxorubicin and gemcitabine, liposomal delivery of paclitaxel does not lead to a better outcome, as was evidenced by a study in a rat prostate cancer xenograft model. Here, efficient tumor inhibition by liposomal paclitaxel was observed at the cost of severe weight loss [26], indicative of excessive systemic toxicity. It may therefore be doubtful whether or not liposomal delivery will increase the therapeutic index of paclitaxel in advanced prostate cancer. In the attempts to further enhance the efficacy of liposomal anticancer drug targeting, two approaches deserve attention: combination therapy and active targeting. Combination therapy of liposomal doxorubicin with radiation [19] or low frequency ultrasound [22] enhanced the antitumor efficacy compared to liposomal doxorubicin alone. In addition, ultrasound was shown to enhance the penetration of released doxorubicin throughout the prostate xenograft, thereby also reaching tumor cells further removed from the blood vessels [23].
580
J. Kroon et al. / Cancer Treatment Reviews 40 (2014) 578–584
Fig. 2. Enhanced permeability and retention (EPR) effect and different modes of liposomal drug delivery. Tumors often display a chaotic and highly permeable vasculature as a result of angiogenic and vascular permeability factors (e.g. VEGF). The long circulation time of PEG-liposomes allows enhanced extravasation of liposomes into the tumor microenvironment. In addition, the lack of proper lymphatic drainage system further contributes to the EPR effect. This so-called passive targeting represents a major targeting principle for liposomes (upper left panel). Active targeting involves a ligand bound to the outer surface of liposomes that selectively target receptors/ligands overexpressed on the tumor cells (upper right panel) or the (a)cellular tumor microenvironment (lower left panel). Following binding to the receptor, internalization via receptor-mediated endocytosis can take place.
Active targeting of receptors on tumor cells and on cancerassociated cells has been pursued, as both tumor and stromal cells may have distinct cellular characteristics which enable selective targeting. In a prostate cancer xenograft model, active targeting of liposomal doxorubicin with an anasamide-PEG derivate to sigma receptors (overexpressed in prostate cancer-derived cell lines [30]) displayed improved antitumor efficacy compared to passively targeted doxorubicin, while free doxorubicin treatment was associated with severe systemic toxicity and treatment-related death [20]. Another active targeting approach focused on fibroblast growth factor receptors (FGFRs), frequently overexpressed in tumor cells and tumor-associated vasculature. In a TRAMP-C1 xenograft model, active FGF-based liposomal delivery of doxorubicin led to a massive reduction in tumor growth and prolonged survival when compared with passively targeted doxorubicin and free doxorubicin [21]. It is unclear to what extent the enhanced antitumor effects were mediated by direct (tumor), indirect (supportive stroma) or combined effects on FGFR-expressing cells. Furthermore, active targeting of tumor vasculature with aspargine-glycine-argenine-(NGR)-targeted liposomes has been explored for prostate carcinoma. NGR selectively binds a tumor endotheliumspecific CD13 isoform and displays a high binding capacity to cultured human vascular endothelial cells (HUVEC) in vitro. Active, NGR-based targeting of doxorubicin induced a dose-dependent inhibition of prostate tumor growth (1–6 mg/kg/week) but was not compared to passively targeted liposomes [31].
In addition to chemotherapeutics, bisphosphonates provide a group of antiresorptive drugs clinically relevant for the treatment of prostate cancer patients with metastatic bone disease. Bisphosphonates home to bone very efficiently due to high affinity for hydroxyapatite which is abundantly present in the calcified bone matrix. At this site, osteoclastic bone resorption is inhibited and for this reason bisphosphonates are widely used in the clinic to prevent tumor-induced bone loss [32]. Several studies highlight the depleting effect of free and liposomal bisphosphonates on tumor-associated macrophages (TAMs) from the tumor microenvironment [33–35]. TAMs are involved in tumor-associated inflammation by secretion of a wide range of cytokines and other inflammatory factors including VEGF, EGF and MMP-9, and they contribute to tumor progression, invasion and angiogenesis [32]. Thus, liposomal targeting of bisphosphonates provides a potential approach to dampen tumor-associated inflammation and tumor progression. Indeed, intravenous injection of liposomal bisphosphonate zoledronic acid resulted in decreased levels of TAMs, reduced angiogenesis and inhibition of prostate xenograft growth [36,37]. In metastatic xenograft models, liposomal delivery of another bisphosphonate, clodronate, led to inhibited metastatic growth and reduced numbers of bone metastases which were accounted to a reduction in TAMs [38,39], reduced levels of inflammatory cytokine IL-6 [39] and a reduction of osteoclast activity [39]. Besides therapeutic potential of bisphosphonates, they can also be used as active targeting devices to selectively deliver anticancer
J. Kroon et al. / Cancer Treatment Reviews 40 (2014) 578–584
drugs to bone metastases. As mentioned earlier, hydroxyapatite is abundantly exposed in the microenvironment of bone metastases leading to enhanced binding by bisphosphonate structures. Indeed, liposomes with a bisphosphonate-moiety display efficient binding to hydroxyapatite in vitro [40–42]. However, hydroxyapatite binding was decreased at increasing serum levels, pointing to competition between serum proteins and bisphosphonate-decorated liposomes [42]. Despite serum competition, it was recently confirmed that bisphosphonate-decorated liposomes display in vivo affinity for collagen/hydroxyapatite scaffolds transplanted in rats [42]. These findings indicate that bisphosphonate-decorated liposomes may provide a means to target anticancer drugs to bone, but active delivery of anticancer drugs has not yet been substantiated and the approach warrants further investigation. Finally, liposomal delivery of antisense oligonucleotides, which inhibit the translation of target messenger RNAs, was evaluated. Antisense oligonucleotides against nucleic acids coding for oncogenic proteins may block the production of pivotal proteins for tumor growth. Using such an approach, Bcl-2 provides a promising target since it inhibits apoptosis and is associated with therapy resistance [43]. Targeted knockdown of Bcl-2 may lead to apoptosis induction or sensitization in tumor cells. Interestingly, intravenous administration of PEGylated cationic liposomes containing Bcl-2 antisense RNA resulted in a dose-dependent inhibition of prostate cancer xenograft growth [44]. These findings indicate successful knockdown of Bcl-2 in vivo, though the intra-tumoral proteins levels of Bcl-2 were not reported [44]. In a similar way, liposomes were used to selectively knock down PKN3, Raf-1 and TMPRSS2/ERG; proteins associated with prostate cancer growth [45–48]. As such, intravenous liposomal administration of siPKN3 led to a significant decrease in tumor size, as well as a strong reduction in the number of affected lymph nodes but, unfortunately, also downregulated PKN3 in healthy tissues [46]. This may point at suboptimal tumor-specificity of the liposomal system. Liposomal delivery of Raf antisense oligonucleotides led to a 50% knockdown of Raf-1 in tumor tissues, which resulted in an enhanced antitumor activity of docetaxel, cisplatin, epirubicin and mitoxantrone on prostate cancer xenografts [48]. This indicates that liposomal siRNA-mediated protein silencing can sensitize prostate xenografts to chemotherapeutics. Another study monitored the effect of liposomal delivery of si-RNA targeted against the TMPRSS2/ERG fusion gene which revealed a significantly inhibition of subcutaneous and intraprostatic xenograft growth while no toxicity was observed [49]. Active targeting of prostate cancer cells to selectively deliver siRNA was explored in a prostate xenograft model, in which potent in vivo knockdown of target Plk-1 was achieved using PSA-responsive, prostate-specific membrane antigen (PSMA)-targeted liposomes, subsequently leading to decreased tumor growth [50]. These multifunctional liposomes offer enhanced selectivity for prostate cancer cells as both PSA and PSMA should be present to facilitate receptor-mediated endocytosis. As can be deducted from the examples above, targeting proteins that are involved in growth and survival of prostate cancer cells may represent a viable treatment approach. In addition, proteins that are involved in the interaction between tumor cells, stromal cells and the extracellular matrix may provide interesting targets. An important protein involved in this communication is av-integrin. It was shown that intra-tumoral injection of liposome-encapsulated av-integrin-siRNA resulted in potent in vivo knockdown and consequently hampered intra-osseous growth of prostate tumor cells [51]. However, intra-tumoral injection is less relevant from a clinical perspective since metastases often present themselves at poorly accessible sites. Targeted delivery of av-integrinsiRNA after systemic administration was not explored in this study [51].
581
Clinical studies With promising preclinical research results with liposomal doxorubicin in prostate cancer models, and the clinical approval of PEGylated and non-PEGylated liposomal doxorubicin in other cancer types, it comes as no surprise that clinical trials with liposomal doxorubicin have also been carried out in CRPC. Indeed, several phase I and phase II trials have been performed evaluating treatment with non-PEGylated [52–54] and PEGylated [55–58] liposomal doxorubicin, either as a monotherapy or in combination with docetaxel [59]. In these studies, serum levels of PSA were used as a read out and a decline of at least 50% was classified as a clinical response. In studies focusing on monotherapy with liposomal doxorubicin, it was notable that patients treated with PEGylated liposomal doxorubicin seemed to respond better compared to patients treated with non-PEGylated liposomal doxorubicin. For example, in the study of McMenemin et al., treatment with PEGylated liposomal doxorubicin leads to a clinical response in 4 out of 14 patients with hormone refractory prostate carcinoma and bone metastases. In another study by Flaherty and coworkers no clinical responses were seen in nine hormone refractory prostate cancer patients treated with non-PEGylated liposomal doxorubicin [52]. Taken together, at least 50% reduction in PSA levels was observed in 18/88 patients (20%, range 11–28%) versus 10/77 patients (13%, range 0–15%) upon treatment with PEGylated and non-PEGylated liposomal doxorubicin, respectively. Of note, different dosing regimens were used in different studies, in which infrequent treatment with high-dose PEGylated liposomal doxorubicin (every 3 or 4 weeks) [55–58] resulted in PSA responses while frequent lowdose treatment (every week) did not [56]. This is exemplified by a phase II trial in which 50 mg/m2 every 4 weeks led to substantial PSA decreases whereas 25 mg/m2 every 2 weeks did not [56]. Even with the most effective dosing schedule of PEGylated doxorubicin, however, only a small proportion of the patient population shows an antitumor response (ranging from 11% to 28%). As synergistic effects of doxorubicin and docetaxel were described in human prostate cancer cells in vitro [60], combination treatment of liposomal doxorubicin with (non-liposomal) docetaxel was evaluated, which shows a P50% decline in PSA level in 50% of the patients with prostate cancer [59]. Treatment with docetaxel alone, however, already results in a PSA decline in 45– 48% of the patients [16], so it is doubtful if addition of liposomal doxorubicin adds much value. In addition to clinical responses, treatment-associated toxicities were monitored. Compared to free doxorubicin, liposomal encapsulation of doxorubicin resulted in reduced cardiotoxicity and less severe myelosuppression, but led to increased dose-limiting skin and mucosal toxicities [55,61] including hand-food syndrome and stomatitis [55,62,63]. This shift in safety profile for liposomal doxorubicin seems favorable, as skin toxicities, unlike cardiotoxicity, are not life threatening and manageable [63]. Treatment with liposomal doxorubicin also infrequently presented with some grade III toxicities including neutropenia [53,54,57,59], leukopenia [56], anemia [59] and thrombocytopenia [58,59]. Nevertheless, for the majority of patients liposomal doxorubicin was well-tolerated, even in combination with docetaxel [59].
Conclusions This review aims to provide a summary of the studies done so far with liposomal drug delivery to prostate carcinoma and its bone metastases. Different potential anticancer drugs were studied including several chemotherapeutics, bisphosphonates and antisense nucleotides (Table 1). In general, passive liposomal targeting of chemotherapeutic doxorubicin displays increased antitumor
582
J. Kroon et al. / Cancer Treatment Reviews 40 (2014) 578–584
Table 1 Overview of preclinical studies on liposomal drug targeting in prostate carcinoma models. Antitumor drug
Liposome composition
Dose & administration
Mean liposome size
Passive/active targeting
In vivo model: animals, cells, tumor inoculation site
References
Doxorubicin
DSPE, DSPC, DSPEPEG2000, CHOL (e.g. Caelyx) PC, CHOL, DSPE-PEGSP2-AA DOTAP, CHOL, tbFGF DSPE, DSPC, DSPE-PEG2000, CHOL (e.g. Caelyx) DEPC, DSPC, DSPE-PEG, CHOL PC, DSPE-PEG-NGR, CHOL PC, CHOL, no PEG PC, CHOL, no PEG Not shown DSPC, CHOL, no PEG
3.5 mg/kg, i.v.
96 nm
Passive
Balb-c nu/nu, human primary cells, s.c.
[19]
7.5 mg/kg/week, i.v.
Not shown
Active, anisamide
[20]
5 mg/kg/2x week, i.v. 3.5 mg/kg, i.v.
162 nm 85 nm
Active, tbFGF Passive
Female athymic nude mice, Du145, s.c. C57BL/6 J, TRAMP-C1, s.c. Balb-c nu/nu, human primary cells, s.c.
16 mg/kg, i.v.
90 nm
Passive
Female Balb-c nu/nu, PC-3, s.c.
[23]
1–6 mg/kg/week, i.v.
Not shown
Active, NGR
[31]
6–8 mg/kg/week, i.v. 8 mg/kg/week, i.v. 5 mg/kg/4 times in 8 days, i.v. 0.5–2 mg/kg/week, i.v.
Not shown 36 nm Not shown Not shown
Passive Passive Passive Passive
Male athymic nude mice, PC-3, s.c. SCID, Du145/PC-3, s.c. SCID, LNCaP, intraprostatic Copenhagen rats, MatLu, s.c. Swiss mice nu/nu, PC-3, s.c.
10–20 lg, i.v.
265 nm, 331 nm Not shown
Passive
Clodronate Bcl-2 siRNA
PC, CHOL, no PEG PC, CHOL, PEG-CLZ
Not shown 90 nm
Passive Passive
PKN3 siRNA
DPhyPE, DSPE-PEG
3x every 5 days, i.p. 1–10 mg/mL, i.v.; 0.1 mg/two 5 day cycles, s.c. 2.8 mg/kg, i.v.
CD-1 nu/nu, PC-3, intramuscular Balb-c nu/nu, HARA-B, intracardiac NCI-nu, PC-3, intraosseous Balb-c nu/nu, PC-3, s.c.
[36–37]
Clodronate
PC, DSPE-PEG2000, CHOL PC, CHOL, no PEG
118 nm
Passive
[46]
Raf antisense
DDAB, PC, CHOL, no PEG DOTAP, DOPC
i.v.
Not shown
Passive
NMRI nu/nu, PC-3, intraprostatic Male athymic nu/nu, PC-3, s.c.
Twice weekly, 150 lg/kg, i.v.
65 nm
Passive
SCID, VCaP, s.c., intraprostatic
[49]
1.5 mg/kg/every 2 days, i.v.
208 nm
Active, Folate
Balc-c nu/nu, 22Rv1, s.c.
[50]
1 lg, i.t.
Not shown
Passive
Balb-c nu/nu, PC-3, intraosseous/s.c.
[51]
Doxorubicin Doxorubicin Doxorubicin
Doxorubicin Doxorubicin Gemcitabine Gemcitabine Paclitaxel Imatinibmitoxantrone Zoledronic acid
TMPRSS2/ERG siRNA PLK-1 siRNA
SPC, DSPE-PEG2000, DSPE-PEG2000-ACPP, DSPE-PEG5000-Folate, CHOL,DC-CHOL av-Integrin siRNA DPPC, DPE, DPPE, PEG
Every 3 days, s.c.
Passive
[21] [22]
[24] [25] [26] [27]
[38] [39] [44]
[47–48]
Abbreviations: siRNA, small interfering RNA; i.v., intravenous; s.c., subcutaneous; i.t., intratumoral; CHOL = cholesterol; PC = phosphocholine; DOTAP = dioleoyl trimethylammonium propane; DDAB = dimethyldioctadecylammoniumbromide; SPC = soybean phosphatidylcholine; DOPC = 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine.
efficacy compared to free doxorubicin, as well as a decrease of side effects. This antitumor effect can potentially be further enhanced by combination therapy or active targeting. Here, targeting of both tumor and stromal cells offers a valuable approach, as exemplified by FGFR-targeted doxorubicin liposomes that show a highly potent antitumor response in a preclinical study. Amongst the performed passive targeting studies, liposomal delivery of gemcitabine seems highly promising as it allows a massive dose reduction while yielding similar antitumor effects as a 45-fold higher free gemcitabine dosage. Liposomal gemcitabine almost completely arrests prostate cancer xenograft growth whereas liposomal doxorubicin only seems to slow it down. Based on this, further investigation on targeted delivery of gemcitabine in preclinical prostate cancer models is warranted. It is important to note, though, that most studies use subcutaneous tumor models to monitor prostate cancer growth in vivo. Although these studies can be useful, the models may not be representative of drug delivery to the relevant biological site, e.g. (bone) metastatic tumor, as tumor microenvironment, vascularization and other properties may differ between subcutaneous and orthotopically implanted tumors. Hence, the use of more clinically relevant in vivo models is key, specifically models that mimic metastatic tumor growth (especially in the bone), as they represent the clinical stage of advanced, incurable, prostate cancer. Indeed, intrabone and intracardiac tumor inoculation, which appear most useful in this regard, were sporadically used in literature as models
for experimentally-induced prostate cancer metastases to evaluate liposomal targeting approaches. The bone/bone-marrow microenvironment, the main site of prostate cancer metastasis, has unique features that can be exploited to selectively deliver anticancer drugs to bone metastases. As such, bisphosphonate-coated liposomes provide a promising, yet underexplored, platform with effective targeting affinity for active bone surfaces (hydroxyapatite), which is abundantly exposed in the local bone metastatic environment. At this site, tumor-associated macrophages promote tumor growth by direct release of growth and inflammatory factors (EGF, VEGF, IL-10, IL12, TNF-a, amongst others) [64,65] while osteoclasts mediate bone resorption, which leads to the release of bone-matrix bound growth factors (for example, TGF-b) [66]. In a broader perspective, we believe that targeting the supportive bone stroma has clear therapeutic potential, as exemplified by the preclinical studies that show that depletion of tumor-associated macrophages by liposomal delivery of bisphosphonates effectively inhibits bone metastatic growth. Although preclinical studies with a range of anticancer drugs show the potential of liposomal drug delivery in prostate cancer treatment, passive targeting studies were only performed with four chemotherapeutic agents so far while only one (doxorubicin) was evaluated in an active targeting approach. Also, despite these and other promising preclinical studies with a range of anticancer drugs, only liposomal doxorubicin has been tested in phase I and II
583
J. Kroon et al. / Cancer Treatment Reviews 40 (2014) 578–584 Table 2 Overview of clinical studies on liposomal drug targeting for prostate cancer treatment. Regimen
PSA P 50% decline Toxicities
Therapeutic
PEGylated? Dose intensity (/week)
Liposomal doxorubicin
No
Liposomal doxorubicin
No
10–20 mg/m2 10–20 mg/m2 weekly for 0/9 (0%) 4 weeks out of a 6-week cycle 2 2 50 mg/m every 4 weeks 2/14 (14%) 12.5 mg/m
Liposomal doxorubicin Liposomal doxorubicin
No Yes
25 mg/m2 15 mg/m2
Liposomal doxorubicin
Yes
12.5 mg/m2
Liposomal doxorubicin
Yes
12.5 mg/m2
Liposomal doxorubicin Yes Liposomal doxorubicin, docetaxel Yes
25 mg/m2 45 mg/m2 60 mg/m2 25 mg/m2 50 mg/m2
weekly every 3 weeks or every 4 weeks every 2 weeks or every 4 weeks
50 mg/m2 every 4 weeks
40 mg/m2 every 4 weeks 10 mg/m2 6–16 mg/m2, 6–16 mg/m2, 20–35 mg/m2 20–35 mg/m2 every week for 3 weeks out of a 4-week cycle
trials for the treatment of prostate cancer (Table 2). Hence, we believe that many challenges remain in the field of liposomal drug delivery for prostate cancer. For the development of such targeting approaches, it is of pivotal importance to exploit prostate tumor and tumor environmental specific characteristics which will likely result in the identification of numerous potential new targets, for which novel liposomal anticancer drugs can be designed and developed. Ultimately, this may lead to the development of highly efficient and specific liposomal drug carriers for the treatment of advanced prostate cancer. Conflict of interest statement The authors declare no conflict of interest. Acknowledgements This study was supported by a grant from the NanoNextNL Drug Delivery Programme (3-D), project number 03D.01. References [1] Lammers T, Kiessling F, Hennink WE, Storm G. Drug targeting to tumors: principles, pitfalls and (pre-) clinical progress. J Control Release 2012 Jul 20;161(2):175–87. PubMed PMID: 21945285. [2] Arias JL. Liposomes in drug delivery: a patent review (2007–present). Expert Opin Ther Pat. 2013. [3] Slingerland M, Guchelaar HJ, Gelderblom H. Liposomal drug formulations in cancer therapy: 15 years along the road. Drug Discov Today 2012;17(3– 4):160–6. PubMed PMID: 21983329. [4] Iyer AK, Singh A, Ganta S, Amiji MM. Role of integrated cancer nanomedicine in overcoming drug resistance. Adv Drug Deliv Rev 2013. PubMed PMID: 23880506. [5] Patterson SG, Wei S, Chen X, Sallman DA, Gilvary DL, Zhong B, et al. Novel role of Stat1 in the development of docetaxel resistance in prostate tumor cells. Oncogene 2006;25(45):6113–22. PubMed PMID: 16652143. [6] de Barros AL, Mota L, Soares DC, de Souza CM, Cassali GD, Oliveira MC, et al. Long-circulating, pH-sensitive liposomes versus long-circulating, non-pHsensitive liposomes as a delivery system for tumor identification. J Biomed Nanotechnol 2013;9(9):1636–43. PubMed PMID: 23980511. [7] Kaasgaard T, Andresen TL. Liposomal cancer therapy: exploiting tumor characteristics. Expert Opin Drug Deliv 2010;7(2):225–43. PubMed PMID: 20095944. [8] Sawant RR, Tochilin VP. Challenges in development of targeted liposomal therapeutics. AAPS J 2012;14(2):303–15. Pubmed Central PMCID: 3326155. [9] Fenske DB, Chonn A, Cullis PR. Liposomal nanomedicines: an emerging field. Toxicol Pathol 2008;36(1):21–9. PubMed PMID: 18337218. [10] Maurer N, Fenske DB, Cullis PR. Developments in liposomal drug delivery systems. Expert opin Biol Ther 2001;1(6):923–47. PubMed PMID: 11728226. [11] Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol 2007;2(12):751–60. PubMed PMID: 18654426.
8/54 (15%) 3/15 (20%) 8/31 (26%)
4/14 (28%) 3/28 (11%) 12/24 (50%)
References
No Grade III or IV
[52]
Grade III neutropenia, mucositis and hand-food syndrome Grade III neutropenia Grade III/IV stomatitis and hand-foot syndrome Grade III/IV tachycardia, hepatic toxicity and hemoglobin toxicity Grade III/IV neutropenia, mucositis and skin toxicity Grade III thrombocytopenia Grade III hematological toxicity, mucositis and ashenia
[53] [54] [55] [56]
[57] [58] [59]
[12] Yuan F, Dellian M, Fukumura D, Leunig M, Berk DA, Torchilin VP, et al. Vascular permeability in a human tumor xenograft: molecular size dependence and cutoff size. Cancer Res 1995;55(17):3752–6. PubMed PMID: 7641188. [13] Hashizume H, Baluk P, Morikawa S, McLean JW, Thurston G, Roberge S, et al. Openings between defective endothelial cells explain tumor vessel leakiness. Am J Pathol 2000;156(4):1363–80. Pubmed Central PMCID: 1876882. [14] Katsogiannou M, Peng L, Catapano CV, Rocchi P. Active-targeted nanotherapy strategies for prostate cancer. Curr Cancer Drug Targets 2011;11(8):954–65. PubMed PMID: 21861840. [15] Kirpotin DB, Drummond DC, Shao Y, Shalaby MR, Hong K, Nielsen UB, et al. Antibody targeting of long-circulating lipidic nanoparticles does not increase tumor localization but does increase internalization in animal models. Cancer Res 2006;66(13):6732–40. PubMed PMID: 16818648. [16] Tannock IF, de Wit R, Berry WR, Horti J, Pluzanska A, Chi KN, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med 2004;351(15):1502–12. PubMed PMID: 15470213. [17] Carles J, Castellano D, Climent MA, Maroto P, Medina R, Alcaraz A. Castrationresistant metastatic prostate cancer: current status and treatment possibilities. Clin Transl Oncol 2012;14(3):169–76. PubMed PMID: 22374419. [18] Deeken JF, Slack R, Weiss GJ, Ramanathan RK, Pishvaian MJ, Hwang J, et al. A phase I study of liposomal-encapsulated docetaxel (LE-DT) in patients with advanced solid tumor malignancies. Cancer Chemother Pharmacol 2013;71(3):627–33. PubMed PMID: 23274395. [19] Hagtvet E, Roe K, Olsen DR. Liposomal doxorubicin improves radiotherapy response in hypoxic prostate cancer xenografts. Radiat Oncol 2011;6:135. PubMed PMID: 21981945. Pubmed Central PMCID: 3193805. [20] Banerjee R, Tyagi P, Li S, Huang L. Anisamide-targeted stealth liposomes: a potent carrier for targeting doxorubicin to human prostate cancer cells. Int J Cancer 2004;112(4):693–700. PubMed PMID: 15382053. [21] Chen X, Wang X, Wang Y, Yang L, Hu J, Xiao W, et al. Improved tumor-targeting drug delivery and therapeutic efficacy by cationic liposome modified with truncated bFGF peptide. J Control Release 2010;145(1):17–25. PubMed PMID: 20307599. [22] Hagtvet E, Evjen TJ, Olsen DR, Fossheim SL, Nilssen EA. Ultrasound enhanced antitumor activity of liposomal doxorubicin in mice. J Drug Target 2011;19(8):701–8. PubMed PMID: 21524240. [23] Eggen S, Afadzi M, Nilssen EA, Haugstad SB, Angelsen B, Davies Cde L. Ultrasound improves the uptake and distribution of liposomal Doxorubicin in prostate cancer xenografts. Ultrasound Med Biol 2013;39(7):1255–66. PubMed PMID: 23643054. [24] Jantscheff P, Esser N, Graeser R, Ziroli V, Kluth J, Unger C, et al. Liposomal gemcitabine (GemLip)-efficient drug against hormone-refractory Du145 and PC-3 prostate cancer xenografts. Prostate 2009;69(11):1151–63. PubMed PMID: 19399788. [25] Jantscheff P, Ziroli V, Esser N, Graeser R, Kluth J, Sukolinskaya A, et al. Antimetastatic effects of liposomal gemcitabine in a human orthotopic LNCaP prostate cancer xenograft model. Clin Exp Metastasis 2009;26(8):981–92. PubMed PMID: 19784785. [26] Bode C, Trojan L, Weiss C, Kraenzlin B, Michaelis U, Teifel M, et al. Paclitaxel encapsulated in cationic liposomes: a new option for neovascular targeting for the treatment of prostate cancer. Oncol Rep 2009;22(2):321–6. PubMed PMID: 19578772. [27] Pinto AC, Moreira JN, Simoes S. Liposomal imatinib-mitoxantrone combination: formulation development and therapeutic evaluation in an animal model of prostate cancer. Prostate 2011;71(1):81–90. PubMed PMID: 20607721. [28] Smith LA, Cornelius VR, Plummer CJ, Levitt G, Verrill M, Canney P, et al. Cardiotoxicity of anthracycline agents for the treatment of cancer: systematic review and meta-analysis of randomised controlled trials. BMC Cancer 2010;10:337. PubMed PMID: 20587042. Pubmed Central PMCID: 2907344.
584
J. Kroon et al. / Cancer Treatment Reviews 40 (2014) 578–584
[29] Chang HI, Yeh MK. Clinical development of liposome-based drugs: formulation, characterization, and therapeutic efficacy. Int J Nanomedicine 2012;7:49–60. PubMed PMID: 22275822. Pubmed Central PMCID: 3260950. [30] Vilner BJ, John CS, Bowen WD. Sigma-1 and sigma-2 receptors are expressed in a wide variety of human and rodent tumor cell lines. Cancer Res 1995;55(2):408–13. PubMed PMID: 7812973. [31] Garde SV, Forte AJ, Ge M, Lepekhin EA, Panchal CJ, Rabbani SA, et al. Binding and internalization of NGR-peptide-targeted liposomal doxorubicin (TVTDOX) in CD13-expressing cells and its antitumor effects. Anticancer Drugs 2007;18(10):1189–200. PubMed PMID: 17893520. [32] Rogers TL, Holen I. Tumor macrophages as potential targets of bisphosphonates. J Tansl Med 2011;9:177. PubMed PMID: 22005011. Pubmed Central PMCID: 3215187. [33] Miselis NR, Wu ZJ, Van Rooijen N, Kane AB. Targeting tumor-associated macrophages in an orthotopic murine model of diffuse malignant mesothelioma. Mol Cancer Ther 2008;7(4):788–99. PubMed PMID: 18375821. [34] Banciu M, Metselaar JM, Schiffelers RM, Storm G. Antitumor activity of liposomal prednisolone phosphate depends on the presence of functional tumor-associated macrophages in tumor tissue. Neoplasia 2008;10(2):108–17. PubMed PMID: 18283332. Pubmed Central PMCID: 2244685. [35] Roelofs AJ, Thompson K, Ebetino FH, Rogers MJ, Coxon FP. Bisphosphonates: molecular mechanisms of action and effects on bone cells, monocytes and macrophages. Curr Pharm Des 2010;16(27):2950–60. PubMed PMID: 20722616. [36] Marra M, Salzano G, Leonetti C, Tassone P, Scarsella M, Zappavinga S, et al. New self-assembly nanoparticles and stealth liposomes for the delivery of zoledronic acid: a comparative study. Biotechnol Adv 2012;30(1):302–9. PubMed PMID: 21741464. [37] Marra M, Salzano G, Leonetti C, Tassone P, Scarsella M, Zappavigna S, et al. Nanotechnologies to use bisphosphonates as potent anticancer agents: the effects of zoledronic acid encapsulated into liposomes. Nanomedicine. 2011;7(6):955–64. PubMed PMID: 21453789. [38] Hiraoka K, Zenmyo M, Watari K, Iguchi H, Fotovati A, Kimura YN, et al. Inhibition of bone and muscle metastases of lung cancer cells by a decrease in the number of monocytes/macrophages. Cancer Sci 2008;99(8):1595–602. PubMed PMID: 18754872. [39] Kim SW, Kim JS, Papadopoulos J, Choi HJ, He J, Maya M, et al. Consistent interactions between tumor cell IL-6 and macrophage TNF-alpha enhance the growth of human prostate cancer cells in the bone of nude mouse. Int Immunopharmacol 2011;11(7):862–72. PubMed PMID: 21251905. Pubmed Central PMCID: 3086935. [40] Hengst V, Oussoren C, Kissel T, Storm G. Bone targeting potential of bisphosphonate-targeted liposomes. Preparation, characterization and hydroxyapatite binding in vitro. Int J Pharm 2007;331(2):224–7. PubMed PMID: 17150316. [41] Anada T, Takeda Y, Honda Y, Sakurai K, Suzuki O. Synthesis of calcium phosphate-binding liposome for drug delivery. Bioorg Med Chem Lett 2009;19(15):4148–50. PubMed PMID: 19523821. [42] Wang G, Mostafa NZ, Incani V, Kucharski C, Uludag H. Bisphosphonatedecorated lipid nanoparticles designed as drug carriers for bone diseases. J Biomed Mater Res A. 2012;100(3):684–93. PubMed PMID: 22213565. [43] Reed JC. Dysregulation of apoptosis in cancer. J Clin Oncol 1999;17(9):2941–53. PubMed PMID: 10561374. [44] Sonoke S, Ueda T, Fujiwara K, Sato Y, Takagaki K, Hirabayashi K, et al. Tumor regression in mice by delivery of Bcl-2 small interfering RNA with pegylated cationic liposomes. Cancer Res 2008;68(21):8843–51. PubMed PMID: 18974128. [45] Leenders F, Mopert K, Schmiedeknecht A, Santel A, Czauderna F, Aleku M, et al. PKN3 is required for malignant prostate cell growth downstream of activated PI 3-kinase. EMBO J 2004;23(16):3303–13. PubMed PMID: 15282551. Pubmed Central PMCID: 514518. [46] Aleku M, Schulz P, Keil O, Santel A, Schaeper U, Dieckhoff B, et al. Atu027, a liposomal small interfering RNA formulation targeting protein kinase N3, inhibits cancer progression. Cancer Res 2008;68(23):9788–98. PubMed PMID: 19047158. [47] Pal A, Ahmad A, Khan S, Sakabe I, Zhang C, Kasid UN, et al. Systemic delivery of RafsiRNA using cationic cardiolipin liposomes silences Raf-1 expression and inhibits tumor growth in xenograft model of human prostate cancer. Int J Oncol 2005;26(4):1087–91. PubMed PMID: 15754006.
[48] Pei J, Zhang C, Gokhale PC, Rahman A, Dritschilo A, Ahmad I, et al. Combination with liposome-entrapped, ends-modified raf antisense oligonucleotide (LErafAON) improves the anti-tumor efficacies of cisplatin, epirubicin, mitoxantrone, docetaxel and gemcitabine. Anticancer Drugs 2004;15(3):243–53. PubMed PMID: 15014358. [49] Shao L, Tekedereli I, Wang J, Yuca E, Tsang S, Sood A, et al. Highly specific targeting of the TMPRSS2/ERG fusion gene using liposomal nanovectors. Clin Cancer Res 2012;18(24):6648–57. PubMed PMID: 23052253. Pubmed Central PMCID: 3525716. [50] Xiang B, Dong DW, Shi NQ, Gao W, Yang ZZ, Cui Y, et al. PSA-responsive and PSMA-mediated multifunctional liposomes for targeted therapy of prostate cancer. Biomaterials 2013;34(28):6976–91. PubMed PMID: 23777916. [51] Bisanz K, Yu J, Edlund M, Spohn B, Hung MC, Chung LW, et al. Targeting ECMintegrin interaction with liposome-encapsulated small interfering RNAs inhibits the growth of human prostate cancer in a bone xenograft imaging model. Mol Ther 2005;12(4):634–43. PubMed PMID: 16039164. [52] Flaherty KT, Malkowicz SB, Vaughn DJ. Phase I study of weekly liposomeencapsulated doxorubicin in patients with advanced, androgen-independent prostate cancer. Am J Clin Oncol 2004;27(2):136–9. PubMed PMID: 15057152. [53] Harris KA, Harney E, Small EJ. Liposomal doxorubicin for the treatment of hormone-refractory prostate cancer. Clin Prostate Cancer 2002;1(1):37–41. PubMed PMID: 15046711. [54] Montanari M, Fabbri F, Rondini E, Frassineti GL, Mattioli R, Carloni S, et al. Phase II trial of non-pegylated liposomal doxorubicin and low-dose prednisone in second-line chemotherapy for hormone-refractory prostate cancer. Tumori 2012 Nov;98(6):696–701. PubMed PMID: 23389354. [55] Hubert A, Lyass O, Pode D, Gabizon A. Doxil (Caelyx): an exploratory study with pharmacokinetics in patients with hormone-refractory prostate cancer. Anticancer Drugs 2000;11(2):123–7. PubMed PMID: 10789595. [56] Heidenreich A, Sommer F, Ohlmann CH, Schrader AJ, Olbert P, Goecke J, et al. Prospective randomized phase II trial of pegylated doxorubicin in the management of symptomatic hormone-refractory prostate carcinoma. Cancer 2004;101(5):948–56. PubMed PMID: 15329902. [57] McMenemin R, Macdonald G, Moffat L, Bissett D. A phase II study of caelyx (liposomal doxorubicin) in metastatic carcinoma of the prostate: tolerability and efficacy modification by liposomal encapsulation. Invest New Drugs 2002;20(3):331–7. PubMed PMID: 12201496. [58] Fossa SD, Vaage S, Letocha H, Iversen J, Risberg T, Johannessen DC, et al. Liposomal doxorubicin (Caelyx) in symptomatic androgen-independent prostate cancer (AIPC)–delayed response and flare phenomenon should be considered. Scand J Urol Nephrol 2002;36(1):34–9. PubMed PMID: 12002355. [59] Kouroussis C, Androulakis N, Vamvakas L, Kalykaki A, Spiridonakou S, Kentepozidis N, et al. Phase I study of weekly docetaxel and liposomal doxorubicin in patients with advanced solid tumors. Oncology 2005;69(3):202–7. PubMed PMID: 16127289. [60] Budman DR, Calabro A, Kreis W. Synergistic and antagonistic combinations of drugs in human prostate cancer cell lines in vitro. Anticancer Drugs 2002;13(10):1011–6. PubMed PMID: 12439335. [61] Lotem M, Hubert A, Lyass O, Goldenhersh MA, Ingber A, Peretz T, et al. Skin toxic effects of polyethylene glycol-coated liposomal doxorubicin. Arch Dermatol 2000;136(12):1475–80. PubMed PMID: 11115157. [62] Uziely B, Jeffers S, Isacson R, Kutsch K, Wei-Tsao D, Yehoshua Z, et al. Liposomal doxorubicin: antitumor activity and unique toxicities during two complementary phase I studies. J Clin Oncol 1995;13(7):1777–85. PubMed PMID: 7602367. [63] O’Brien ME, Wigler N, Inbar M, Rosso R, Grischke E, Santoro A, et al. Reduced cardiotoxicity and comparable efficacy in a phase III trial of pegylated liposomal doxorubicin HCl (CAELYX/Doxil) versus conventional doxorubicin for first-line treatment of metastatic breast cancer. Ann Oncol 2004;15(3):440–9. PubMed PMID: 14998846. [64] Liu CY, Xu JY, Shi XY, Huang W, Ruan TY, Xie P, et al. M2-polarized tumorassociated macrophages promoted epithelial-mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway. Lab Invest 2013;93(7):844–54. PubMed PMID: 23752129. [65] Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell 2010;141(1):39–51. PubMed PMID: 20371344. [66] Sturge J, Caley MP, Waxman J. Bone metastasis in prostate cancer: emerging therapeutic strategies. Nat Rev Clin Oncol 2011;8(6):357–68. PubMed PMID: 21556025.