Review

Proteasome inhibitor patents (2010 -- present) Rainer Metcalf, Latanya M Scott, Kenyon G Daniel & Q Ping Dou† †

Wayne State University, Karmanos Cancer Institute, Detroit, MI, USA

1.

Overview

2.

Current proteasome inhibitors: novel chemical entities created specifically for inhibiting the

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

proteasome 3.

Combination therapy: known or novel compounds used in combination with other known drugs for proteasome inhibition and synergy of therapeutic effect

4.

Expert opinion

Introduction: Over the past 3 years, numerous patents and patent applications have been submitted and published involving compounds designed to inhibit the proteasome. Proteasome inhibition has been of great interest in cancer research since disruption of proteolysis leads to a significant buildup of cytotoxic proteins and activation of apoptotic pathways, particularly in rapidly proliferating cells. The current standards in proteasome inhibition are the only FDA-approved inhibitors, bortezomib and carfilzomib. Although these drugs are quite effective in treating multiple myeloma and other blood tumors, there are shortcomings, including toxicities and resistance. Most of the current patents attempt to improve on existing compounds, by increasing bioavailability and selectivity, while attempting to reduce toxicity. A general categorization of similar compounds was employed to evaluate and compare drug design strategies. Areas covered: This review focuses on novel compounds and subsequent analogs developed for proteasome inhibition, used in preventing and treating human cancers. A comprehensive description and categorization of patents related to each type of compound and its derivatives, as well as their uses and efficacies as anticancer agents is included. A review of combination therapy patents has also been included. Expert opinion: Although there are many diverse chemical scaffolds being published, there are few patented proteasome inhibitors whose method of inhibition is genuinely novel. Most patents utilize a destructive chemical warhead to attack the catalytic threonine residue of the proteasome active sites. Few patents try to depart from this, emphasizing the need for developing new mechanisms of action and specific targeting. Keywords: anticancer therapy, bortezomib, carfilzomib, clinical trials, drug discovery, patents, polyphenols, proteasome inhibitors Expert Opin. Ther. Patents (2014) 24(4):369-382

1.

Overview

Protein turnover is an essential part of amino acid metabolism in which cells constantly synthesize and degrade proteins. The rate of expression and degradation of proteins is critical for the proper regulation of metabolic pathways. The proteasome is a nearly universal cellular component and integral for this fundamental biological process [1,2]. The 26S mammalian proteasome is a massive 2400 kDa molecule comprising a 20S core particle (CP) and one or two 19S 18-subunit regulatory particle (RP) [3]. The RP, or PA700 cap, also incorporates specific recognition sites to which ubiquitin chains can bind [4]. The CP is a 700 kDa, barrel-shaped structure composed of four six-subunit rings [5]. The a subunits of the outer rings are largely structural in function and appear to regulate the formation and stabilization of either the 26S proteasome or 19S immunoproteasome [6]. Proteolysis occurs at the six protease active sites of the inner ring b subunits oriented toward the lumen

10.1517/13543776.2014.877444 © 2014 Informa UK, Ltd. ISSN 1354-3776, e-ISSN 1744-7674 All rights reserved: reproduction in whole or in part not permitted

369

R. Metcalf et al.

Article highlights. . . .

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

.

Proteasome inhibition remains an effective technique in cancer therapy. A few recent proteasome inhibitor patents are genuinely novel in their method of inhibition. Most patents attempt to utilize epigallocatechin gallate or a peptidomimetic moiety coupled with a chemically reactive warhead. None of the currently published compounds appear to have the optimal combination of potency, toxicity and bioavailability.

This box summarizes key points contained in the article.

of the proteasome [3]. These active sites comprise a catalytic region and multiple recognition regions [7]. The catalytic regions are similar and incorporate a catalytic threonine 1 (Thr 1) residue to execute the hydrolysis of the protein [3]. Discretionary binding of specific residue sequences is accomplished by the recognition regions of the active sites [8]. These sites are designated as the b1/PRE3peptidylglutamyl peptide caspase-like recognition site (PGPH), the b2/PUP1 trypsinlike recognition site (T-L) and the b5/PRE2 chymotrypsinlike recognition site (CT-L) [9]. Other structural variants of the proteasome have been discovered and they exhibit slightly altered functions of the b subunits and regulatory caps. The immunoproteasome consists of smaller 11S RPs and a modified 20S CP containing ib1, ib2 and ib5 subunits and structural isoforms of the constitutive b1, b2 and b5 subunits, respectively [10]. The thymoproteasome is another proteasomal polymorphism more recently elucidated, which incorporates the ib1 and ib2 immunoproteasome subunits but integrates a constitutively differing tb5 subunit [11]. Disruption of proteolysis can lead to significant buildup of cytotoxic proteins and activation of apoptotic pathways, particularly in rapidly proliferating cells [12]. For this reason, proteasome inhibition has been of great interest in cancer research [13]. Further studies into the use of proteasome inhibitors on oncogenic cell lines have revealed that they may prevent angiogenesis and metastasis and increase susceptibility to apoptosis, while quiescent cells may be preserved to some extent [14]. Possible therapies for other diseases and disorders, such as neurodegenerative disorders [15], cardiac disease [16,17] and organ transplant rejection [18], are being researched. The patent search strategy was carried out with focus on patents or applications from the past 3 years that cover new chemical entities, or new methods of using natural products, known chemical entities, or any combinations of the aforementioned as inhibitors of proteasome activity. Additionally, the scope of utility of these patents was not limited to only cover cancer therapy but included patents that teach proteasome inhibition as a method of treating other diseases, such as immunological disorders or viral infections. The search 370

was initiated broadly to capture the most patents and applications filed between 1 January 2010 and 30 September 2013, using the Delphion patent database from Thomson Reuters. This search focused on finding representative classes of new chemical entities being claimed as novel proteasome inhibitors.

Current proteasome inhibitors: novel chemical entities created specifically for inhibiting the proteasome

2.

Proteasome inhibitors can be categorized into several different chemical classifications. A recent review article written by our group outlined a generalized list of current proteasome inhibitors based on chemical structure and reaction mechanism [19]. Many patents tend to circulate around existing structures, seeking to improve potency and selectivity. These analogs possess a similar pharmacophore comprising two key elements: a peptide portion that selectively binds to the recognition binding pocket of the proteasome with high affinity and a chemical moiety specifically designed to interact with the catalytic Thr 1 residue to irreversibly inhibit protease activity [20]. Common functional groups that are exploited to react with the catalytic Thr 1 residue are aldehydes, vinyl sulfones, boronates, epoxyketones and b-lactones. Boron-containing compounds Boron-containing inhibitors capitalize on highly reactive boron-containing ‘warheads’ to create a covalent ligature to a protein. Therefore, these types of molecules are generally noncompetitive and extremely potent, with IC50 values often in the nanomolar and picomolar range. One of the most well-known proteasome inhibitors, bortezomib (Figure 1-1), is of this type, utilizing a boronic acid moiety to perform a nucleophilic attack on the CT-L catalytic Thr 1 residue. This mechanism is a frequently adopted design strategy with attempts geared mainly to decrease side effects. Although bortezomib is a potent and effective proteasome inhibitor, it has been found to have several and often severe side effects, including fatigue, nausea, sensory neuropathy and adverse cardiovascular effects [21,22]. Additionally, the development of bortezomib resistance in patients and preliminary studies showing limited effect on solid tumors are being investigated [23-26]. Li et al. developed multiple compounds directed primarily at alleviating the side effects of bortezomib. The most active compound, 6f (Figure 1-2), displayed a CT-L IC50 of 0.079 ± 0.011 µM in biochemical assays, as well as an IC50 of 3.630 ± 1.669 µM in the PGPH-active site [27]. It also exhibited greater efficacy in cellular assays than bortezomib at 0.5 µM across multiple cell lines: HL-60 human leukemia with 87% inhibition, BGC-823 human gastric cancer with 95% inhibition, bel-7402 human hepatocarcinoma with 94% inhibition and Kb human nasopharyngeal carcinoma 2.1

Expert Opin. Ther. Patents (2014) 24(4)

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

Proteasome inhibitor patents (2010 -- present)

1

2

3

4

5

6

7

8

Figure 1. Boron-containing compounds are shown.

with 96% inhibition. Further, in vivo testing showed a 55% tumor inhibition [27]. Unfortunately, a 63% survival rate resonated as on par with the toxicity of bortezomib [27]. Compound 6g (Figure 1-3), in contrast, presented an efficacy comparable to bortezomib in biochemical and cellular assays, but with a 100% survival rate in rat models [27].

An interesting approach to the use of a chemical warhead by utilizing a type of protecting group on the warhead can be illustrated in the patent by Bernardini et al. [28]. This protecting group serves two purposes: one being that it masks the polarity of the highly polar boronic acid moiety, thereby increasing bioavailability, and two, it attempts to decrease

Expert Opin. Ther. Patents (2014) 24(4)

371

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

R. Metcalf et al.

1

2

3

4

5

Figure 2. Epoxyketone-containing compounds are shown.

the reactivity of the warhead until binding, conceivably reducing off-target effects. The patent outlined a large array of example compounds, but the general structure shown in Figure 1-4 characterizes the basic form of the protecting group on the boronic acid warhead. The group did a simple high-throughput screening to ascertain IC50 and EC50 values for each compound [28]. Compounds with the general form in Figure 1-4 displayed biochemical assay IC50 values of < 10 nM and EC50 values of < 200 nM in Molt 4 cells [28]. The compound shown in Figure 1-5 is a specific example, representative of the general form (Figure 1-4). Other recent patents on bortezomib analogs include compounds I-20 (Figure 1-6) [29], with a reported IC50 372

of < 50 nM and a list of compounds with the general structure shown in Figure 1-7 by Shenk et al. [30]. Another patent of note is on distinct formulations for stabilizing bortezomib in a liquid form suitable for injection [31]. While this patent may not seek claim for a specific compound, it does show alternative patent strategies in the realm of drug design. Epoxyketones Epoxyketones garnered significant interest after the introduction of carfilzomib (Figure 2-1) as a safer, more specific and more effective proteasome inhibitor than bortezomib [32]. Carfilzomib was designed as an analog to epoxomicin based on its in vivo antitumor activity [33]. The epoxyketone 2.2

Expert Opin. Ther. Patents (2014) 24(4)

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

Proteasome inhibitor patents (2010 -- present)

pharmacophore is the main chemical moiety developed to attack the CT-L catalytic Thr 1 residue by forming a dual covalent morpholino adduct [34]. Compound 1 (Figure 2-2) was patented by Kirk and Jiang and showed that dosages of 30 -- 40 mg/kg reduce the number of visible metastatic tumors in lungs by ~ 50% [35]. Shenk et al. applied for a patent on compound 14 (Figure 23) specifically as a potential treatment for rheumatoid arthritis. Their work showed that dosages at 6 mg/kg reduced arthritis symptom severity in rat models by 50% [30,36]. Smyth et al. published multiple patents focusing solely on the structure and synthesis of compounds that emphasize a diversity of peptide mimetics employing an epoxyketone warhead [37-39]. The general structure is shown in Figure 2-4. Most proteasome inhibitors target the hydrophobic CT-L site since the majority of pharmaceutically viable compounds must be relatively hydrophobic themselves. This leaves the other sites severely underresearched as targets. Kisselev et al. developed compounds with the intent of specifically inhibiting the T-L active site [40]. The compound NC-022 (Figure 2-5) displayed preferential binding to the T-L site with an IC50 of 38 nM [40]. Other analogs portrayed similar potencies; however, NC-022 was the most cell-permeable compound and, thus, held the greater potential as an applicable therapeutic [40]. The authors also found that vinyl sulfones tended to be more potent and more selective for T-L than epoxyketones with same sequence and showed that T-L inhibition sensitizes cells to CT-L inhibitors, leading to possible combination therapies mentioned in later sections [40]. Despite the success of carfilzomib, epoxyketones capitalize on the same basic mechanism as bortezomib and share many of the common patient inconveniences and side effects. Although Carfilzomib demonstrated a 22.9% overall response rate in clinical studies and showed reduced incidence of peripheral neuropathies compared to bortezomib [9,41], carfilzomib still presented some side effects including pneumonia, acute renal failure, pyrexia, congestive heart failure, cytopenia, fatigue, nausea and dyspnea [32,41]. Continued research and improvements on this family of compounds are necessary to yield more safe and effective therapeutic options. Green tea-derived polyphenolic compounds Epigallocatechin gallate (EGCG) (Figure 3-1) has gained considerable interest in cancer treatment due to evidence of its ability to inhibit growth of several types of cancers [42-49]. Other studies have also shown EGCG to have certain selective binding to the proteasome, making it a potential model for designing novel proteasome inhibitors [50]. EGCG belongs to a class of natural products known as flavonoids, commonly found in green tea and other edible plants [51]. The EGCG analogs, compounds 5 and 7 (Figure 3-2 and 3-3, respectively), studied by Chan et al. show promise in the treatment of multiple myeloma (MM) ARP cells with in vitro tests displaying 70% growth inhibition at 50 µM for 2.3

compound 5 and 95% growth inhibition for compound 7 at the same concentration [52]. Another patent by the same author endeavored to increase the bioavailability of EGCG by increasing the hydrophobicity of the compounds and decreasing the rate of metabolism by introducing protecting groups [53]. By acetylating all of the hydroxyl groups on EGCG, the authors were able to create a prodrug (compound 1, Figure 3-4) that exhibited slower metabolic degradation and greater cell permeability, while retaining the potency of EGCG [53]. Patents for synthetic polyphenolic compounds developed by our group show interesting improvements in efficacy by changing the stereochemistry of natural products. By switching the chirality of the natural compounds, (--)-EGCG (Figure 3-1) and (--)-GCG (Figure 3-5), to the opposite plane polarized forms, (+)-EGCG (Figure 3-6) and (+)-GCG (Figure 3-7), the authors were able to substantially increase potency from IC50 values of 205 nM for (--)-EGCG to 170 nM for (+)-EGCG, and 610 nM for (--)-GCG to 270 nM for (+)-GCG [54]. Unmodified flavonoids are generally weak inhibitors, with common IC50 values in the range of many micromolars. However this property, coupled with their low toxicity and antioxidant activities, allows flavonoid analogs to fulfill a unique niche as low-risk agents that could be incorporated into diet as part of a preventative strategy for disease. Metal complexes Metal complexes using gold [55,56], nickel [57,58], zinc [57,58] and to a greater degree copper [59-63], have received significant attention due to their unique pharmacophore. Copper complexes have been viewed with particular promise as potential proteasome inhibitors, in addition to a nascent strategy in cancer therapy, after studies showed that cancer cells accumulate more copper than healthy cells [59-63]. 8--hydroxyquinoline (8OHQ) (Figure 4-1) has been well known as a monoprotic bidentate chelating agent for some time [64], although its potential as a proteasome inhibitor was only recently elucidated when our group found that the 8OHQ analog, clioquinol (Figure 4-2), inhibited the CT-L activity of the proteasome, blocked proliferation and induced apoptosis in breast cancer cells, while remaining nontoxic to noncancerous breast cells [60]. Since then, there have been several novel inhibitor designs incorporating the basic 8OHQ structure. Recent analogs, 5-amino-8-hydroxyquinoline (5AHQ) (Figure 4-3), of 8OHQ by Schimmer include an additional amino group and has similar IC50 values as 8OHQ [65]. In vivo assays of 5AHQ against human leukemia displayed modest IC50 values of 3.7 ± 0.3 µM. Sheshbaradaran exploited the preformed gallium metal complex, tris(8quinolinolato)gallium(III) (Figure 4-4), at 2.5 -- 5 µM in combination with 5 nM concentration of bortezomib to show a near 100% growth inhibition of lung cancer tissue [66]. 2.4

Expert Opin. Ther. Patents (2014) 24(4)

373

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

R. Metcalf et al.

1

2

3

4

5

6

7

Figure 3. Green tea-derived polyphenolic compounds are shown.

374

Expert Opin. Ther. Patents (2014) 24(4)

Proteasome inhibitor patents (2010 -- present)

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

1

indicate a strong potential for a viable treatment of various cancers [73-76]. Natural products isolated from marine organisms have served as an exceptional source for unique lead compounds in drug design [77,78]; therefore, it is not surprising to see a representative of this class of compounds showing potential as a prime archetype for a new class of novel proteasome inhibitors. Salinosporamide will likely continue to be of interest to researchers and a robust pharmacophore in future drug design strategies.

2

Unique chemical designs Whereas many patents utilize previously discovered structures as a pharmacophore for developing novel compounds, there have been some recent publications employing entirely original structures. The apparent majority of effective proteasome inhibitors are noncompetitive and make use of some sort of ‘warhead’ to covalently modify the active site threonine residues. Competitive proteasome inhibitors rarely achieve activities lower than several micromolars. However, Lawrence et al. discovered an interesting compound, PI-1833 (Figure 6-1), reporting an IC50 of 630 ± 350 nM. The group also tested multiple analogs with some (PI-1840, Figure 6-2) achieving reported values of 32 ± 0.15 nM, alluding to the possibility of having found the first competitive proteasome inhibitors comparable to bortezomib [79]. The same group also discovered another unique compound, HLM-008182 (Figure 6-3), with a reported IC50 of 650 ± 40 nM [80]. Another innovative patent by Bachovchin et al. [81] capitalizes on the overexpression of fibroblast activation protein (FAP) in stromal fibroblasts of epithelial cancers [82]. FAP binds to a specific peptide sequence attached to a cytotoxic moiety; Figure 6-4 is an example of one such peptide sequence. The enzyme cleaves the compound to release the active moiety only when present in cells with sufficient expression of FAP [81]. While the active moiety may not necessarily be a proteasome inhibitor, the patent includes many of the aforementioned warheads utilized as the active group, R, in Figure 6-5. The patent focuses primarily on the mechanism of FAP-activated compounds, allotting to the possible incorporation of any cytotoxic moiety for the treatment of epithelial carcinomas. Although the novelty of this patent is not technically in proteasome inhibition, its inclusion in this review is significant due to the patent’s unique attempt to specifically deliver a proteasome inhibitor to certain cancer cells. Even though these compounds have only recently been discovered and are a long way from the clinical use, further investigation and design improvement will no doubt yield compounds with considerable potential as therapeutic agents. In order to surmount the obstacles faced by these promising leads, an ongoing research effort is essential to understand how these compounds achieve such high potencies and possibly elucidate new pharmacophores for effective competitive proteasome inhibition. 2.6

3

4

Figure 4. Metal complexes are shown.

Salinosporamide analogs The marine natural product, salinosporamide A (Figure 5-1), has recently received considerable notice mainly due to its remarkable potency as a proteasome inhibitor with IC50 values often reaching the low picomolar range. The g-lactam--blactone bicyclic core of salinosporamide A is the fundamental structure and also functions as a proteasome inhibitor by covalently modifying active site threonine residues [67-69]. The replacement of the chlorine group on salinosporamide A to a hydroxyl group (Figure 5-2) led to a substantial decrease in cytotoxicity, 38 ± 4 µM compared to 9.8 ± 3 nM, while retaining potency, CT-L IC50 of 14 ± 1.5 nM compared to 2.5 ± 1.2 nM, and increasing selectivity for the CT-L active site [70]. Various compounds with the general structure in Figure 5-3 were patented by Ling et al. [71]. The patent included a method for synthesizing the natural product and the structures of any intermediates formed in the process. The authors also compared the differences in inhibition between production through fermentation and synthetic manufacturing [71]. The synthetic form, being more pure, exhibited a slightly better CT-L IC50 of 3.2 nM and 96% inhibition of the 20S proteasome in RPMI 8826 cells at 10 nM concentrations [71]. Although the natural form of salinosporamide A cannot be patented, Fenical et al. have applied for a patent on multiple stereoisoforms of salinosporamide [72] and early clinical trials of the natural compound, clinically named marizomib, 2.5

Expert Opin. Ther. Patents (2014) 24(4)

375

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

R. Metcalf et al.

1

2

3

Figure 5. Salinosporamide analogs are shown.

3. Combination therapy: known or novel compounds used in combination with other known drugs for proteasome inhibition and synergy of therapeutic effect

Biochemical pathways in cells are seldom isolated. Fully inhibiting one enzyme may deplete a specific product or stress the cell but will rarely shutdown an entire pathway in vivo. Other intersecting metabolic routes can assist in replenishing products and may even completely overcome the induced ‘roadblock’ [83-87]. Additionally, being tiny crucibles of strained genetic evolution, cancerous tissue is constantly battling to survive utilizing the sheer brute force of mathematical chance. Cancer cells require only a single chance adaption to a drug to gain resistance [88,89]. The method of combination therapy seeks to address these problems, since a tumor cell is far less likely to gain resistance to multiple drugs before dying. Further, different drugs can act to inhibit alternate pathways or sensitize the cell to other drugs, leading to synergistic effects. Fertig et al. designed an anti-CD20 antibody, a humanized B-Ly1 antibody, to be used alone and in combination with several proteasome inhibitors, including Bortezomib, 376

rituximab and cyclophosphamide [90]. Mice xenographs of SU-DHL-4 non-Hodgkin’s lymphoma cells showed 87% inhibition with the antibody alone [90]. Although this therapy was effective by itself, combination therapy with bortezomib resulted in lymphoma regression and complete tumor remission with no regrowth in four of nine animals [90]. Forster capitalized on patenting the usage of several types of glucocorticoids in conjunction with proteasome inhibitors to mitigate brain or spinal cord damage following an ischemic event [91]. By stabilizing the blood--brain barrier with glucocorticoids to diminish tissue edema and administering a proteasome inhibitor in an attempt to prevent the degradation of the inhibitory protein IkB-a, which downregulates NF-kB, a necrosis factor, neuronal death can be abated long enough until the tissue can be reperfused or oxygenated [91]. Delanzomib (Figure 1-8) is another compound discovered some time ago with a similar pharmacophore as bortezomib [92]. This drug has recently gone through Phase I clinical trials for treatment of MM and advanced solid tumors [93]. Delanzomib does appear to alleviate some of the peripheral neuropathy seen in bortezomib treatments but it still suffers from many of the same side effects, albeit to a lesser degree [93]. Although delanzomib is currently being researched as a possible cancer treatment, Ruggeri and Seavey have already submitted a patent for its use as a potential treatment of lupus [94]. Studies have found that the administration of delanzomib improved survival and reduced lupus nephritis in MRL/lpr and BWF1 mouse models [95]. Some patents mentioned previously that contained additional outlines for possible combination therapies are 5AHQ (Figure 4-3), which when combined with 10 nM concentration of bortezomib exhibited a synergistic effect across multiple cell lines, including MM, leukemia and solid tumors [65]. The T-L inhibitor, NC-022 (Figure 2-4), developed by Kisselev et al. exhibited a strong synergistic effect with other CT-L inhibitors [40]. When combined with 33 nM bortezomib, an 81 ± 19% growth inhibition of MM cells compared to 47 ± 13% with bortezomib alone at the same concentration was noticed [40]. The authors also tested the compound with a minor 3.7 nM concentration of carfilzomib to achieve a 64 ± 15% growth inhibition in MM cells, a substantial contrast to the near negligible 1 ± 9% growth inhibition with the same concentration of carfilzomib alone [40]. A patent can be obtained solely on a method of using a certain chemical entity. However, if there exists another patent on the structure of that chemical entity, the applicant of the method--of-use patent would need the permission of the other patent holder to be able to legally practice the method-of-use invention. When this occurs, the other patent is said to ‘dominate’ the first patent. With this in mind, patents that solely cover the methods of using a combination of chemical entities tend to be lower on patent hierarchy. Although easily overshadowed, the value of a pure method of use patent becomes apparent if no one can obtain a patent

Expert Opin. Ther. Patents (2014) 24(4)

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

Proteasome inhibitor patents (2010 -- present)

1

2

3

4

5

Figure 6. Unique chemical designs are shown.

for the chemical entity itself. This can manifest when a chemical entity is found to occur in nature. Although they can be discovered or isolated, natural products themselves are not patentable subject material. The caveat lies in that distinct methods of using natural products are patentable subject material. In this case, there is more of a level playing field, where a chemical entity structure patent cannot dominate a method of use patent. 4.

Expert opinion

In general, a person has a right to patent an invention in USA as long as the invention is not an abstract idea, a law of nature, or naturally occurring, and is useful, novel and non-obvious to one of ordinary skill in the art. What that equates to in layman’s terms is that an invention must be conceived of and made by humans, must have some useful purpose, must have never been known or publicly described before seeking a patent and is not an obvious invention to one with general and basic skill and knowledge in the field. Many factors come into play when deciding to file a patent application for a particular invention, with the potential value of the

invention’s utility often being a major factor. In the pharmaceutical field, one can think of a chemical entity’s value as hierarchical; patents on the structure of new chemical entities are often considered the most valuable, because they carry the most weight in terms of excluding others from making or practicing a patented invention. No one else can use that chemical entity for any purpose without the holder’s permission throughout the duration of the patent term, because they own the rights to make and use the structural matter. The fact that most of the patents and applications that we uncovered in our searches include, in some way, claiming new chemical entities themselves is reflective of the prime value of this type of patent position. There are over 10 structurally distinct classes of proteasome inhibitors, signifying a rich diversity of chemical space. This is important for the novelty of a patent; however, nearly all of the aforementioned chemical entities can be categorized into two general types based on the mechanism of action: a chemical warhead affixed to peptide for recognition or copying EGCG to make a competitive inhibitor. Most patents make use of a chemical warhead and a peptidomimetic moiety to selectively bind to the CT-L active site, ignoring the other

Expert Opin. Ther. Patents (2014) 24(4)

377

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

R. Metcalf et al.

two sites completely, and covalently attack the catalytic threonine. Covalent inhibitors are chemically reactive, tending to produce toxic metabolites and have severe off-target reactions. As a result, competitive inhibitors may be more desirable when tissue targeting is not considered, since current competitive inhibitors tend to have less severe side effects. This trend is not absolute, however, as the mechanisms of competitive inhibitors are often less understood compared to noncompetitive inhibitors, possess poorer potency and many have a similar number of off-target interactions. Based on the patent landscape, research into competitive proteasome inhibitors is limited and innovation in this field is also something to be desired. Repeatedly, others attempting to make a true competitive inhibitor synthesize EGCG analogs based on the information of EGCG--proteasome b5 interactions. The bioavailability of EGCG and analogs poses pharmacological challenges but provides a unique role in the realm of prevention vis-a`-vis diet for this class of compounds. This argument of novelty is semantic but it does attempt to expose entrenched methods of drug design. Although the practice of improving a previously discovered pharmacophore does yield results, it is essential to focus on developing new mechanisms of action and, more importantly, specific targeting. Specific targeting of cancer cells has remained an elusive goal. Due to this, most drugs attempt to take advantage of the fact that oncogenic tissues tend to be more susceptible to cytotoxic effects than normal healthy tissue. Therefore, introducing compounds that cause global toxicity should, theoretically, kill the cancer before killing the patient. This concept has yielded less than desirable results and has often led to the question of which is worse, the cure or the disease. Current attempts at proteasome inhibitors are no different. Whereas global inhibition of the proteasome has been shown to have limited toxic effects on healthy tissue, this does not render healthy tissue immune to proteasome inhibition or resistant to other off-target effects that may arise due to the presence of a chemically reactive warhead. Further, bortezomib might have some off-targets, which are associated with the emergence of peripheral neuropathy. There has been evidence in clinical trials that bortezomib may have other proteasome-independent mechanisms of action in addition to proteasome inhibition [96-103]. These articles show alternate pathways being activated or downregulated, leading to cytotoxicity and possible causes of adverse drug effects. Analysis of gene expression profiles and single nucleotide polymorphisms (SNPs) of myeloma plasma cells and peripheral blood samples have identified genes and SNPs associated with mitochondrial dysfunction and DNA repair [98]. Whereas these can be expressed through proteasomal pathways, interference with ribosome function, essential DNA damage pathways and dysregulation of many other metabolic processes can be attributed to other off-target interactions [104,105]. Previous studies have indicated that bortezomib-induced neurodegeneration in vitro occurs exclusively by way of a proteasomeindependent mechanism and that bortezomib inhibits several 378

non-proteasomal peptidase targets in vitro and in vivo [106,107]. Another gene expression profiling study found that the cytotoxic effects of bortezomib were not p27-dependent and confirmed that bortezomib increases the expression of genes associated with endoplasmic reticulum stress, whereas proteasome subunit knockdown alone does not [108]. Consistently, carfizolmib, which is more specific to inhibit the CT activity than bortezomib, has been shown to have decreased levels of peripheral neuropathy in myeloma patients, compared to bortezomib. Along this line, any compound utilizing a chemical warhead will suffer from these off-target interactions to some extent. Although a peptide mimic attached to the chemical warhead increases selectivity, the reactivity of these warheads is independent of binding with intended targets and it is likely that the warhead will react with other targets in similar chemical environments. If a drug can properly protect the warhead until binding, global inhibition is still a viable strategy so long as the side effects can be suitably mitigated. On the other hand, toxic compounds can be utilized with great efficacy if they can be delivered exclusively to cancer cells. Either way, any truly effective cancer therapy must involve a solution for specific targeting and/or exclusive delivery to cancer tissue. Over the past decade, proteasome inhibition has emerged as an effective method for treating MM and other blood cancers, in which bortezomib plays an important role. However, some limitations were identified in bortezolmib’s use. A prominent factor is adduced from the severe side effects that relegate clinical studies of bortezomib to late stage, terminal cancer patients. In late-stage cancer, treatments utilizing proteasome inhibition are ineffective, since apoptotic machinery in the cancer cell is usually dismantled. Carfilzomib has improved specificity and decreased peripheral neuropathy. It will remain to be further investigated if carfilzomib can assuage some criticisms of proteasome inhibition as a cancer therapy in practical application. Hopefully, researchers can learn from the difficulties of bortezomib and carfilzomib and develop other clinical candidates and therapeutics that can attain the perfect balance of potency, toxicity and bioavailability in the future.

Acknowledgments The authors would like to thank N Shakfeh for her assistance in drawing the chemical structures. R Metcalf and LM Scott have contributed equally to the manuscript.

Declaration of interest This work was supported by the Moffitt Cancer Center Research Institute, the University of South Florida College of Arts and Sciences, Karmanos Cancer Institute and Wayne State University. The authors declare no conflict of interest and have received no payment in preparation of this manuscript.

Expert Opin. Ther. Patents (2014) 24(4)

Proteasome inhibitor patents (2010 -- present)

Bibliography Papers of special note have been highlighted as either of interest () or of considerable interest () to readers. 1.

Rubin DM, Finley D. The proteasome: a protein-degrading organelle? Curr Biol 1995;5:854-4

2.

Baumeister W, Walz J, Proteolysis C. The proteasome: compartmentalizing protease. Cell 1998;92:367-80

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

3.

4.

5.

6.

7.

8.

9.

Groll M, Ditzel L, Lowe J, et al. Structure of 20S proteasome from yeast at 2.4 A resolution. Nature 1997;386(6624):463-71 Elsasser S, Gali RR, Schwickart M, et al. Proteasome subunit Rpn1 binds ubiquitin-like protein domains. Nat Cell Biol 2002;4:725-30 Pickart CM, Cohen RE. Proteasomes and their kin: proteases in the machine age. Nat Rev Mol Cell Biol 2004;5:177-87 Bose S, Stratford F, Broadfoot K, et al. Phosphorylation of 20S proteasome alpha subunit C8 (alpha7) stabilizes the 26S proteasome and plays a role in the regulation of proteasome complexes by gamma-interferon. Biochem J 2004;378:177-84 Borissenko L, Groll M. 20S proteasome and its inhibitors: crystallographic knowledge for drug development. Chem Inform 2007;38:687-717 Tsvetkov L, Nanjundan M, Domino M, et al. The ubiquitin-proteasome system and assays to determine responses to inhibitors. Expert Opin Drug Discov 2010;5:1221-36 Kisselev AF, van der Linden WA, Overkleeft HS. Proteasome inhibitors: an expanding army attacking a unique target. Chem Biol 2012;19:99-115

13.

Rajkumar SV, Richardson PG, Hideshima T, et al. Proteasome inhibition as a novel therapeutic target in human cancer. J Clin Oncol 2005;23:630-9

14.

Almond J, Cohen G. The proteasome: a novel target for cancer chemotherapy. Leukemia 2002;16:433

15.

16.

17.

25.

Kondagunta GV, Drucker B, Schwartz L, et al. Phase II trial of bortezomib for patients with advanced renal cell carcinoma. J Clin Oncol 2004;22:3720-5

26.

Markovic SN, Geyer SM, Dawkins F, et al. A phase II study of bortezomib in the treatment of metastatic malignant melanoma. Cancer 2005;103:2584-9

Paul S. Dysfunction of the ubiquitin--proteasome system in multiple disease conditions: therapeutic approaches. Bioessays 2008;30:1172-84

27.

Li R, Cui J, Zhu Y, et al. Tripetide boronic acid or boronic ester, preparative method and use thereof. Google Patents. US13379142; 2010

Willis MS, Townley-Tilson WD, Kang EY, et al. Sent to destroy: the ubiquitin proteasome system regulates cell signaling and protein quality control in cardiovascular development and disease. Circ Res 2010;106:463-78

28.

Bernardini R, Bernareggi A, Cassara PG, et al. Proteasome inhibitors and methods of using the same. US8058262; 2011 An interesting attempt to decrease toxicity and increase bioavailability by employing a protecting group on the boronic acid moiety.

Yu X, Kem DC. Proteasome inhibition during myocardial infarction. Cardiovasc Res 2010;85:312-20

.

29.

Olhava EJ, Danca MD. Proteasome inhibitors. US13209511; 2011

Flechner SM, Fatica R, Askar M, et al. The role of proteasome inhibition with bortezomib in the treatment of antibodymediated rejection after kidney-only or kidney-combined organ transplantation. Transplantation 2010;90:1486-92

30.

Shenk KD, Paralati F, Bennett MK. Compounds for immunoproteasome inhibition. US0072422; 2013

31.

Soppimath K, Pejaver S, Patel KR, et al. Stable Bortezomib Formulations. US0322763; 2012

19.

Pevzner Y, Metcalf R, Kantor M, et al. Recent advances in proteasome inhibitor discovery. Expert Opin Drug Discov 2013;8:537-68

32.

20.

Kisselev AF, Goldberg AL. Proteasome inhibitors: from research tools to drug candidates. Chem Biol 2001;8:739-58

Fostier K, De Becker A, Schots R. Carfilzomib: a novel treatment in relapsed and refractory multiple myeloma. Oncol Targets Ther 2012;5:237

33.

Kuhn DJ, Chen Q, Voorhees PM, et al. Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitinproteasome pathway, against preclinical models of multiple myeloma. Blood 2007;110:3281-90

34.

Demo SD, Kirk CJ, Aujay MA, et al. Antitumor activity of PR-171, a novel irreversible inhibitor of the proteasome. Cancer Res 2007;67:6383-91

35.

Kirk CJ, Jiang J. Use of Peptide Epoxyketones for Metastasis Suppression. US0035295; 2013

36.

Shenk KD, Parlati F, Zhou HJ, et al. Compounds for enzyme inhibition. US8080576; 2011

37.

Smyth MS, Laidig GJ. Compounds for proteasome enzyme inhibition. US8198270; 2012

18.

21.

Adams J. Potential for proteasome inhibition in the treatment of cancer. Drug Discov Today 2003;8(7):307-15

22.

Zhu Y, Zhao X, Zhu X, et al. Design, synthesis, biological evaluation, and structure- activity relationship (SAR) discussion of dipeptidyl boronate proteasome inhibitors, part I: comprehensive understanding of the SAR of alpha-amino acid boronates. J Med Chem 2009;52:4192-9

10.

Kloetzel PM, Ossendorp F. Proteasome and peptidase function in MHC-class-Imediated antigen presentation. Curr Opin Immunol 2004;16:76-81

11.

Murata S, Takahama Y, Tanaka K. Thymoproteasome: probable role in generating positively selecting peptides. Curr Opin Immunol 2008;20:192-6

23.

An W, Hwang S, Trepel J, et al. Protease inhibitor-induced apoptosis: accumulation of wt p53, p21WAF1/ CIP1, and induction of apoptosis are

Mujtaba T, Dou Q. Advances in the understanding of mechanisms and therapeutic use of bortezomib. Discov Med 2011;12:471-80

24.

Shah MH, Young D, Kindler HL, et al. Phase II study of the proteasome inhibitor bortezomib (PS-341) in patients

12.

with metastatic neuroendocrine tumors. Clin Cancer Res 2004;10:6111-18

independent markers of proteasome inhibition. Leukemia 2000;14:1276

Expert Opin. Ther. Patents (2014) 24(4)

379

R. Metcalf et al.

38.

Smyth MS, Laidig GJ. Compounds for enzyme inhibition. US8207125; 2012

39.

Smyth MS, Laidig GJ. Compounds for enzyme inhibition. US8207127; 2012

40.

Kisselev A, Filippov DV, Overkleeft H. Inhibitors of the trypsin-like site of the proteasome and methods of use thereof. US8455431; 2013 A very insightful paper into the usefulness of T-L active site inhibitors as well as unique compounds with high selectivity for the T-L active site.

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

..

41.

42.

43.

44.

45.

46.

47.

48.

380

US Food and Drug Administration. Approved Drugs - Carfilzomib. 2012. Available from: http://www.fda. gov/Drugs/InformationOnDrugs/ ApprovedDrugs/ucm312945.htm Guo S, Lu J, Subramanian A, et al. Microarray-assisted pathway analysis identifies mitogen-activated protein kinase signaling as a mediator of resistance to the green tea polyphenol epigallocatechin 3-gallate in her-2/ neu--overexpressing breast cancer cells. Cancer Res 2006;66:5322-9

49.

Smith DM, Daniel KG, Wang Z, et al. Docking studies and model development of tea polyphenol proteasome inhibitors: applications to rational drug design. Proteins 2003;54:58-70

61.

Milacic V, Jiao P, Zhang B, et al. Novel 8-hydroxylquinoline analogs induce copper-dependent proteasome inhibition and cell death in human breast cancer cells. Int J Oncol 2009;35:1481-91

50.

Nam S, Smith DM, Dou QP. Ester bond-containing tea polyphenols potently inhibit proteasome activity in vitro and in vivo. J Biol Chem 2001;276:13322-30

62.

51.

Bonfili L, Cecarini V, Amici M, et al. Natural polyphenols as proteasome modulators and their role as anti-cancer compounds. FEBS J 2008;275:5512-26

Hindo SS, Frezza M, Tomco D, et al. Metals in anticancer therapy: copper (II) complexes as inhibitors of the 20S proteasome. Eur J Med Chem 2009;44:4353-61

63.

Zuo J, Bi C, Fan Y, et al. Cellular and computational studies of proteasome inhibition and apoptosis induction in human cancer cells by amino acid schiff base-copper complexes. J Inorg Biochem 2013;118:83-93

64.

Hollingshead RGW. Studies on derivatives of 8-hydroxyquinoline (oxine). The sensitivity and selectivity of 5-nitroso-oxine and 2-methyl-5-nitrosooxine towards the group IIIB metals. Anal Chim Acta 1955;12:401-7

65.

Schimmer AD. Use of 5AHQ and bortezomib for the treatment of hematological diseases. US13203878; 2010

66.

Sheshbaradaran H. Combination therapy with A proteasome inhibitor and a gallium complex. US0096068; 2013

67.

Feling RH, Buchanan GO, Mincer TJ, et al. Salinosporamide A: a highly cytotoxic proteasome inhibitor from a novel microbial source, a marine bacterium of the new genus Salinospora. Angew Chem Int Ed 2003;42:355-7

68.

Fenical W, Jensen PR, Palladino MA, et al. Discovery and development of the anticancer agent salinosporamide A (NPI-0052). Bioorg Med Chem 2009;17:2175-80

69.

Lam KS, Lloyd GK, Neuteboom STC, et al. From natural product to clinical trials: NPI-0052 (salinosporamide A), a marine actinomycete-derived anticancer agent. Chapter 12. Nat Prod Chem Drug Discov 2010;355-73

70.

Macherla VRR, Potts BC, Manam RR, et al. Proteasome inhibitors. US8227503; 2012

71.

Ling T, Macherla VRR, Potts BC, et al. Total synthesis of salinosporamide A and analogs thereof. US8067616; 2011

72.

Fenical WH, Jensen PR, Mincer TJ, et al. Salinosporamides and methods for use thereof. US8217072; 2012

73.

Chauhan D, Singh A, Brahmandam M, et al. Combination of proteasome

52.

Chan TH, Dou QP. Polyphenol compounds for inhibiting proteasome and uses thereof. US12968759; 2010

53.

Chan TH, Lam WH, Chow LMC, et al. (-)-epigallocatechin gallate derivatives for inhibiting proteasome. US8193377; 2012

54.

Dou QP, Chan TH, Smith DM. Polyphenol proteasome inhibitors, synthesis, and methods of use. US13271058; 2011

55.

Sun CL, Yuan JM, Koh WP, et al. Green tea, black tea and breast cancer risk: a meta-analysis of epidemiological studies. Carcinogenesis 2006;27:1310-15

Milacic V, Chen D, Ronconi L, et al. A novel anticancer gold(III) dithiocarbamate compound inhibits the activity of a purified 20S proteasome and 26S proteasome in human breast cancer cell cultures and xenografts. Cancer Res 2006;66:10478-86

Sartippour MR, Heber D, Ma J, et al. Green tea and its catechins inhibit breast cancer xenografts. Nutr Cancer 2001;40:149-56

56.

Landau JM, Wang ZY, Yang GY, et al. Inhibition of spontaneous formation of lung tumors and rhabdomyosarcomas in A/J mice by black and green tea. Carcinogenesis 1998;19:501-7

Milacic V, Dou QP. The tumor proteasome as a novel target for gold (III) complexes: implications for breast cancer therapy. Coord Chem Rev 2009;253:1649-60

57.

Cvek B, Milacic V, Taraba J, et al. Ni (II), Cu (II), and Zn (II) diethyldithiocarbamate complexes show various activities against the proteasome in breast cancer cells. J Med Chem 2008;51:6256-8

58.

Frezza M, Hindo SS, Tomco D, et al. Comparative activities of nickel (II) and zinc (II) complexes of asymmetric [NN O] ligands as 26S proteasome inhibitors. Inorg Chem 2009;48:5928-37

59.

Daniel KG, Gupta P, Harbach RH, et al. Organic copper complexes as a new class of proteasome inhibitors and apoptosis inducers in human cancer cells. Biochem Pharmacol 2004;67:1139-51

Horie N, Hirabayashi N, Takahashi Y, et al. Synergistic effect ofgreen tea catechins on cell growth and apoptosis induction in gastric carcinoma cells. Biol Pharm Bull 2005;28:574-9 Bettuzzi S, Brausi M, Rizzi F, et al. Chemoprevention of human prostate cancer by oral administration of green tea catechins in volunteers with high-grade prostate intraepithelial neoplasia: a preliminary report from a one-year proof-of-principle study. Cancer Res 2006;66:1234-40 Kemberling J, Hampton JA, Keck RW, et al. Inhibition of bladder tumor growth by the green tea derivative epigallocatechin-3-gallate. J Urol 2003;170:773-6

60.

Daniel KG, Chen D, Orlu S, et al. Clioquinol and pyrrolidine dithiocarbamate complex with copper to form proteasome inhibitors and apoptosis inducers in human breast cancer cells. Breast Cancer Res 2005;7:R897-908

Expert Opin. Ther. Patents (2014) 24(4)

Proteasome inhibitor patents (2010 -- present)

inhibitors bortezomib and NPI-0052 trigger in vivo synergistic cytotoxicity in multiple myeloma. Blood 2008;111:1654-64 74.

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

75.

76.

77.

78.

79.

80.

81.

..

82.

83.

Ruiz S, Krupnik Y, Keating M, et al. The proteasome inhibitor NPI-0052 is a more effective inducer of apoptosis than bortezomib in lymphocytes from patients with chronic lymphocytic leukemia. Mol Cancer Ther 2006;5:1836-43 Miller CP, Ban K, Dujka ME, et al. NPI-0052, a novel proteasome inhibitor, induces caspase-8 and ROS-dependent apoptosis alone and in combination with HDAC inhibitors in leukemia cells. Blood 2007;110:267-77 Miller CP, Rudra S, Keating MJ, et al. Caspase-8 dependent histone acetylation by a novel proteasome inhibitor, NPI0052: a mechanism for synergy in leukemia cells. Blood 2009;113:4289-99 Sallam AA, Ramasahayam S, Meyer SA, et al. Design, synthesis, and biological evaluation of dibromotyrosine analogues inspired by marine natural products as inhibitors of human prostate cancer proliferation, invasion, and migration. Bioorg Med Chem 2010;18:7446-57 Mudit M, Khanfar M, Muralidharan A, et al. Discovery, design, and synthesis of anti-metastatic lead phenylmethylene hydantoins inspired by marine natural products. Bioorg Med Chem 2009;17:1731-8

modularaity in metabolic networks. Science 2002;297:1551-5 84.

Jeong H, Tombor B, Albert R, et al. The large-scale organization of metabolic networks. Nature 2000;407:651-4

85.

Barabasi AL, Oltvai ZN. Network biology: understanding the cell’s functional organization. Nat Rev Genet 2004;5:101-13

86.

Ma H, Zeng AP. Reconstruction of metabolic networks from genome data and analysis of their global structure for various organisms. Bioinformatics 2003;19:270-7

87.

Taylor KB. Enzyme kinetics and mechanisms. 2002; Kluwer Academic Publishers, Norwell: MA, USA

88.

Gottesman MM. Mechanisms of cancer drug resistance. Annu Rev Med 2002;53:615-27

89.

Dean M, Fojo T, Bates S. Annual review of medicine. Tumour stem cells and drug resistance. Nat Rev Cancer 2005;5:275-84

90.

Fertig G, Friess T, Klein C, et al. Combination therapy of a type II anti-CD20 antibody with a proteasome inhibitor. US0219549; 2012

91.

Forster C. Means and methods for treating ischemic conditions. US0245078; 2012

92.

Lawrence HR, Sebti SM, Ozcan S, et al. Proteasome chymotrypsin-like inhibition using PI-1833 analogs. WO129564; 2012 Lawrence H, Ge Y, Sebti S, et al. Proteasome inhibitors having chymotrypsin-like activity. WO102286; 2010 Bachovchin W, Lai HS, Poplawski S, et al. FAP-activated proteasome inhibitors for treating solid tumors. WO033396; 2013 An interesting patent on targeting certain types of cancer cells. Scanlan MJ, Raj B, Calvo B, et al. Molecular cloning of fibroblast activation protein alpha, a member of the serine protease family selectively expressed in stromal fibroblasts of epithelial cancers. Proc Natl Acad Sci 1994;91:5657-61 Ravasz E, Somera AL, Mongru DA, et al. Hierarchical organization of

93.

Piva R, Ruggeri B, Williams M, et al. CEP-18770: a novel, orally active proteasome inhibitor with a tumorselective pharmacologic profile competitive with bortezomib. Blood 2008;111:2765-75 Gallerani E, Zucchetti M, Brunelli D, et al. A first in human phase I study of the proteasome inhibitor CEP-18770 in patients with advanced solid tumours and multiple myeloma. Eur J Cancer 2013;49(2):290-6

94.

Ruggeri B, Seavey M. Proteasome inhibitor delanzomib for use in the treatment of lupus. WO119056; 2012

95.

Li R, Yang N, Zhang L, et al. Inhibition of Jak/STAT signaling ameliorates mice experimental nephrotic syndrome. Am J Nephrol 2007;27:580-9

96.

Nikrad M, Johnson T, Puthalalath H, et al. The proteasome inhibitor bortezomib sensitizes cells to killing by death receptor ligand TRAIL via BH3-only proteins Bik and Bim. Mol Cancer Ther 2005;4:443-9

Expert Opin. Ther. Patents (2014) 24(4)

97.

Williams S, Pettaway C, Song R, et al. Differential effects of the proteasome inhibitor bortezomib on apoptosis and angiogenesis in human prostate tumor xenografts. Mol Cancer Ther 2003;2:835-43

98.

Broyl A, Corthals SL, Jongen JL, et al. Mechanisms of peripheral neuropathy associated with bortezomib and vincristine in patients with newly diagnosed multiple myeloma: a prospective analysis of data from the HOVON-65/GMMG-HD4 trial. Lancet Oncol 2010;11:1057-65

99.

Nawrocki ST, Carew JS, Pino MS, et al. Bortezomib sensitizes pancreatic cancer cells to endoplasmic reticulum stressmediated apoptosis. Cancer Res 2005;65:11658-66

100. Lonial S, Waller EK, Richardson PG, et al. Risk factors and kinetics of thrombocytopenia associated with bortezomib for relapsed, refractory multiple myeloma. Blood 2005;106:3777-84 101. Zangari M, Yaccoby S, Cavallo F, et al. Response to bortezomib and activation of osteoblasts in multiple myeloma. Clin Lymphoma Myeloma 2006;7:109-14 102. Takeda K, Mori A, Yanagida M. Identification of genes affecting the toxicity of anti-cancer drug bortezomib by genome-wide screening in S. pombe. PloS One 2011;6:e22021 103. McConkey DJ. A novel role for a familiar protein in apoptosis induced by proteasome inhibition. Cancer Cell 2008;14:1-2 104. Shoshan MC, Linder S. Target specificity and off-target effects as determinants of cancer drug efficacy. Expert Opin Drug Metab Toxicol 2008;4(3):273-80 105. Dick LR, Fleming PE. Building on bortezomib: second-generation proteasome inhibitors as anti-cancer therapy. Drug Discov Today 2010;15:243-9 106. Arastu-Kapur S, Anderl JL, Kraus M, et al. Nonproteasomal targets of the proteasome inhibitors bortezomib and carfilzomib: a link to clinical adverse events. Clin Cancer Res 2011;17:2734-43 107. Chen S, Blank JL, Peters T, et al. Genome-wide siRNA screen for modulators of cell death induced by

381

R. Metcalf et al.

proteasome inhibitor bortezomib. Cancer Res 2010;70:4318-26 108. Nickeleit I, Zender S, Sasse F, et al. Argyrin A reveals a critical role for the tumor suppressor protein p27 in mediating antitumor activities in response to proteasome inhibition. Cancer Cell 2008;14:23-35

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Washington on 08/28/14 For personal use only.

Affiliation Rainer Metcalf1, Latanya M Scott2, Kenyon G Daniel*1,4 & Q Ping Dou†3,4 †,* Authors for correspondence 1 Moffitt Cancer Center, Chemical Biology Core, 12902 Magnolia Dr SRB3, Tampa, FL 33612, USA 2 Moffitt Cancer Center, Office of Technology Management and Commercialization, 12902 Magnolia Dr MRC-TTO Tampa, FL 33612, USA 3 Wayne State University, Hudson Webber Cancer Research Center, The Developmental Therapeutics Program, 4100 John R. Street Room 540.1, Detroit, MI 48201, USA Tel: +1 813 745 5734; E-mail: [email protected] 4 Wayne State University, Karmanos Cancer Institute, 540.1 HWCRC, 4100 John R Road, Detroit, MI 48201, USA Tel: +1 313 576 8301; Fax: +1 313 576 8307; E-mail: [email protected]

382

Expert Opin. Ther. Patents (2014) 24(4)

Proteasome inhibitor patents (2010 - present).

Over the past 3 years, numerous patents and patent applications have been submitted and published involving compounds designed to inhibit the proteaso...
626KB Sizes 0 Downloads 0 Views