A Drug of Such Damned Nature.1 Challenges and ... - ACS Publications

Feb 14, 2017 - landscape and challenges for future Pt development and discuss opportunities ...... The challenge is the need for the newer “me-tooâ€...
0 downloads 0 Views 1MB Size
Subscriber access provided by University of Newcastle, Australia

Perspective

A drug of such damned nature . Challenges and opportunities in translational platinum drug research. Dorian M Cheff, and Matthew D. Hall J. Med. Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jmedchem.6b01351 • Publication Date (Web): 14 Feb 2017 Downloaded from http://pubs.acs.org on February 16, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Journal of Medicinal Chemistry is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

A drug of such damned nature.1 Challenges and opportunities in translational platinum drug research.

Dorian M. Cheff and Matthew D. Hall* NCATS Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States

ACS Paragon Plus Environment

1

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 66

Abstract The platinum-based anti-cancer agents cisplatin, carboplatin and oxaliplatin, represent a spectacular translational science achievement. The basic research observations that led to the discovery of Pt complexes as DNA-binding agents that elicit cell arrest, the preclinical tumor regression studies, and the inorganic medicinal chemistry that led to clinical implementation of effective platinum complexes in the clinic, have fueled multidisciplinary research into platinum-based drugs. While the successes are clear and the research activity continues, a significant window of time has passed since a new Pt drug has been approved for clinical use. Here we assess the current Pt drug landscape, challenges for future Pt development, and discuss opportunities for improving our understanding of Pt drugs that utilize contemporary translational science tools such as chemical biology and real-time imaging. The underexplored spaces may reveal new opportunities for Pt drug development.

ACS Paragon Plus Environment

2

Page 3 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Introduction Platinum stands out among chemotherapeutic classes for its spectacular successes and utility in combination therapy, but also for its inconvenience to the patient and the resistance that can arise. The first report related to the discovery of cisplatin was published over 50 years ago (1965) (see Hoeschele2 for a detailed description of the early experiments). However, as is well reported, it was likely first synthesized by Michele Peyrone around 1840,3 and was critical to Nobel prize-winner Alfred Werner’s work on inorganic isomerism.4

Cisplatin became the first of three platinum-based anticancer agents registered for use with the FDA (1978), followed by carboplatin (1989) and oxaliplatin (2002) (structures shown in Figure 1). Other Pt-based drugs have entered clinical trials, and some are approved for use in specific countries (discussed below). In parallel with this clinical effort, significant basic research has been conducted on Pt-based complexes with a number of aims, including: to understand their mechanism(s) of action; to synthesize and test new Pt complexes as anticancer drug candidates; to characterize the ways in which cells become resistant to Pt drugs; to explore whether other (non-Pt) metal complexes can offer useful biological activity; to study the (bio)chemistry of Pt complexes in the context of reactions with biomolecules; and to characterize the cellular pharmacology and fate of Pt drugs.

This perspective aims to serve as a reflection on the ghosts of platinum research past, present, and future. It is written to focus attention and inspire discussion on the strengths

ACS Paragon Plus Environment

3

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 66

of the field, and the future of Pt research. It was inspired partly by Richard Callaghan’s honest reflections on the relevance of drug transporters in cancer resistance5 entitled ‘Providing a molecular mechanism for P-glycoprotein; why would I bother?’. The corresponding author has spent 15 years studying Pt complexes, over that time transitioning from inorganic medicinal chemistry and biological spectroscopy, to cell biology and mechanisms of resistance to Pt drugs.6-9 The discussion here is written from the perspective of one who has been immersed in the field of Pt drugs, but who, through the natural peregrinations of research, has evolved a view of the field from afar that might offer an external perspective on the strengths and weaknesses of the field. It is also an attempt to be candid. The difference is vast between the honest (and colloquial) posterside conversation at conferences on the state and direction of a field, and the sober sanitized nature of published perspectives that are written to ensure no future reviewer will be offended. In that context, this Perspective aims to be a useful conversation piece.

Like other ‘micro-fields’ of study, the topic of ‘metals-in-medicine’ (or inorganic medicinal chemistry) has been subject to the vicissitudes of biomedical fashions and fads. With each new technology that emerges to improve cancer treatment, the hand-wringing over the relevance of ‘shotgun’ therapeutics begins.10 It seems, anecdotally, that the typical expectation is that the need for Pt drugs will decline as a result of the new (‘better’) technology, followed by the realization that the technology is not the complete solution that everyone anticipated, and finally the clinical demonstration that the new technology works very well in combination with a Pt drug!

ACS Paragon Plus Environment

4

Page 5 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

O H 3N

Cl

H 3N

Pt H 3N

O

H2 N

H 3N

O O

Cisplatin

O

O

O

Pt

Pt Cl

O

Carboplatin

N H2

Oxaliplatin

Figure 1. Structures of cisplatin, carboplatin, and oxaliplatin. In each case the cis nonleaving amines are at the left, and the leaving groups (chloride, bidentate dicarboxylate, or oxalate) are at the right.

The health impact and economic benefit of Pt drugs has been tremendous.11 While these authors do not believe Pt drugs will leave the clinic for many generations to come, the expectation seems to be that ‘shotgun’ therapeutics should be phased out, and this is understandable given the side-effects experienced by patients, and the superiority of targeted drugs against some malignancies in the genomic age.12 From a basic and translational research point of view, new biomedical discoveries with therapeutic implications should be welcomed as a way to empower Pt research and effectiveness in the clinic. A colleague once said that the major referee critique against their rejected grant application was that with the arrival of gene therapy, there would no longer be a need for chemotherapy!

Despite the time that has passed since the discovery of cisplatin, the number of publications per year continues to increase for all three clinical Pt complexes. There are more than 60,000 publications indexed in Pubmed for ‘cisplatin’, and more in 2015 than any other year (>3,200). The same is true for carboplatin (more than 14,000 total, >790 in

ACS Paragon Plus Environment

5

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 66

2015) and oxaliplatin (over 8,000 total, >850 in 2015) (depicted in Figure 2A). It is notable that oxaliplatin surpassed carboplatin in publications in 2015 for the first time. Comparison with other DNA-targeting chemotherapeutic agents developed in the same period (Figure 2B, chlorambucil, mitomycin, mitoxantrone, bleomycin) emphasizes the scientific and medical impact of the ‘platins’. In each case, an initial surge in publications per year occurred, followed by a plateau. In fact, the only ‘shotgun’ chemotherapeutics that appear to come close are doxorubicin (Adriamycin) and the ‘blockbuster’ drug paclitaxel (Taxol), which show similar research attention (Figure 2C). Even a modern ‘blockbuster’ targeted drug such as the Bcr-Abl inhibitor imatinib (Glivec) shows static research output, while the antibody therapeutic trastuzumab (Herceptin), which targets HER2/neu receptor, does display continued growth in research output. The comparisons in publications made here are reflective only of scientific interest, they are not a measure of clinical impact, but among the >80,000 indexed papers on platinum drugs (and likely many thousands more on other Pt experimental therapeutics and compounds) there are a core of innovative papers that set the tone for topics of interest. This includes topics such as structure activity relationships,13 Pt-DNA structures,14 structure-rule-breakers15 such as active trans complexes16, and highly active non-Pt complexes such as osmium,17 to name a few examples. While citation tracking of published papers that pre-exist the internet are not complete, the 10 most highly cited research reports (excluding literature reviews, using Google Scholar18) describing the three clinical Pt drugs all appear to be clinical reports (not shown).

ACS Paragon Plus Environment

6

Page 7 of 66

A.

Publications per year

4000 3000 2000 1000 0 1970

1980

1990

2000

2010

Year Cisplatin

Carboplatin

Oxaliplatin

B. 600

Publications per year

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

400

200

0 1950

1960

1970

1980

1990

2000

2010

Year Chlorambucil

Mitomycin

Mitoxantrone

ACS Paragon Plus Environment

Bleomycin

7

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 66

C.

Figure 2. A. Publications indexed in Pubmed referring to the drugs, cisplatin, carboplatin, or oxaliplatin in the title or abstract, displayed by year. B. As a comparison, publications describing the FDA-registered DNA-damaging agents chlorambucil, mitomycin, mitaxantrone and bleomycin. C. Publications describing ‘blockbuster’ oncology therapeutics paclitaxel, doxorubicin, imatinib and trastuzumab.

Given the remarkable clinical importance of Pt drugs, it is critical to keep in mind the contrasting negative perception of them. Certainly the lay-person’s view of the drug being a toxic ‘heavy metal’ is technically accurate, perhaps the worst press being Lance Armstrong’s inaccurate statement that “for testicular cancer, they treat you with plutonium, one of the harshest chemicals there is,”19 (though the cure itself is testament to the drug’s effectiveness). Armstrong also stated in his autobiography that they gave him “…a cocktail of three different drugs, bleomycin, etoposide, and cisplatin, and they were so toxic that the nurses wore radioactive protection when handling them”20 (emphasis

ACS Paragon Plus Environment

8

Page 9 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

added) – either confusing the radiotherapy arm of treatment, or the personal protective equipment (PPE) worn by nurses to minimize chronic workplace exposure to cytotoxic (and possibly mutagenic) drugs.

The historical clinical literature is peppered with the perspective that cisplatin would not be approved by the FDA were it discovered today. Cisplatin’s clinical usefulness was ultimately rescued by pre-hydration to offset nephrotoxicity and anti-emetics to manage nausea,21 after much experimentation. The challenge of irreversible ototoxicity in patients, which is not a dose-limiting toxicity but results life-changing effects such as deafness and tinnitus, remains unresolved.22 It is interesting to consider just how close cisplatin came to not being registered in spite of its activity.23 The engrossing and lengthy transcript of the Witness Seminar held by the Wellcome Trust Center for the History of Medicine in 200623 contains multiple references by participants reflecting on the promising efficacy of cisplatin against malignancies such as ovarian cancer in early trials in England (circa 1971/2), but also the clinical challenges associated with toxicity. These challenges were a significant topic of discussion at the 1973 Oxford meeting,24 perhaps best captured by Hill who summarized pessimistically that ‘New and better analogs are awaited with great interest,’25 and this consensus led to the desire for less toxic analogs. The Johnson Matthey work on analogs led to the development of carboplatin (initially coded JM-826), but other options were pursued. While not mentioned in the official proceedings publication that followed the 1973 meeting, nor in the Witness Seminar, the popular science writer June Goodfield attended the 1973 meeting, and described the inprogress Pt story in a chapter of her book ‘Cancer Under Siege’27 based on interviews she

ACS Paragon Plus Environment

9

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 66

conducted with researchers at the meeting. The Pt chapter title “I want the platinum blues!” is a direct reference to Barnett Rosenberg’s stated belief during the 1973 meeting that the platinum blue complexes,28 with their lesser nephrotoxicity than cisplatin, would be the ultimate clinical Pt drug!

Nevertheless, cisplatin and carboplatin prevailed. Any reader who has not observed a patient suffering from the specific effects of platinum chemotherapy need to look no further than a social media platform such Twitter29 to read posts by patients. Alongside the aforementioned cisplatin side-effects, oxaliplatin produces peripheral neuropathy that results in cold hypersensitivity, such as shooting pains if a patient’s skin comes into contact with a metal surface,30 as well as peripheral neurotoxicity, often causing paresthesias and numbness. The drugs certainly stay with patients—it has been shown that platinum is detectable in the urine of subjects 8 years following their last Pt chemotherapy round.31 In the veterinary setting, cisplatin also presents benefits and challenges. While cisplatin is a single-agent curative for equine sarcoids (administered directly into the tumor site via needle as an oil suspension)32, 33, all veterinary trainees also learn that ‘cisplatin splats cats’, a reference to the species-specific pulmonary edema and death that precludes its use in felines.34

Intrinsic and acquired resistance to Pt drugs also occurs in patients. Drug resistance is normally studied by culturing a cell line in increasing concentrations of a given drug to force the acquisition of a resistance phenotype. It is recognized that 2-dimensional tissue culture is not the ideal cell model, but it is certainly the easiest model to study). Another

ACS Paragon Plus Environment

10

Page 11 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

weakness in this strategy is that single-agent resistance in cell lines is very different from the combination chemotherapies a tumor is normally exposed to, and therefore develops resistance to. For example, patients diagnosed with ovarian cancer usually undergo surgical resection (debulking), followed by chemotherapy composed of a platinum (cisplatin or carboplatin) and a taxane (paclitaxel or docetaxel) drug, repeated every 21 or 28 days.35, 36 A recent Nature supplement on tackling drug resistance in ovarian cancer singled out platinum as the cause of the resistance problem.37 Despite the fact that the front-line treatment for high-grade serous ovarian carcinoma is a taxane and a platinum, the article’s accompanying video narration states “most die because their cancer develops resistance to the most common form of treatment – platinum based chemotherapy” (i.e., the taxane is not to blame). The article is unfairly titled ”The Problem with Platinum” despite being about ovarian cancer, and looks forward to ‘new approaches (that) promise to break through the platinum barrier.’38 The article does not explain that platinum chemotherapy extends the life of suffers by years, and that it is front-line because no replacement chemotherapy regimen has been demonstrated to be superior. This example reinforces the negative image of platinum chemotherapy among those who peer-review manuscripts on new Pt agents, review Pt grant applications, and read Pt publications. Yet Pt drugs are an essential part of many chemotherapeutic regimens. When asked why they studied cisplatin resistance, a world-known clinician shrugged and said simply, “because it will still be in the clinic in 50 years, so we need to continue to try and understand it.”

The critical path for researchers in experimental therapeutics is to translate discoveries towards the clinic.39 As such, the design, synthesis, and study of experimental Pt

ACS Paragon Plus Environment

11

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 66

complexes are, by definition, translational. The critical question is: how can Pt drugs be relevant to new discoveries in our understanding of cancer, and how can Pt drugs be used in concert with new therapeutic strategies? It is incumbent on metals-in-medicine experts to ask these questions, because there are unique aspects to Pt drugs, and their chemistry, which limit those outside the field to appropriately design experiments using Pt complexes—for this reason, collaboration is key. When the corresponding author recently published a study on the effect of DMSO on the cancer cell line cytotoxicity and mechanism of cell death of Pt drugs, and the extent to which DMSO is used as the solvent for Pt drugs in the literature,40 he was met with some very positive41 and some very negative feedback (personal communication!). The fact that Pt complexes react with DMSO was already well known within the metals-in-medicine community,42 which is dwarfed in size by the oncology community using the compound in experimental studies. The finding that over a third of the oncology and experimental therapeutic literature used DMSO as a solvent for Pt drugs was distressing as the implications for the extant literature were potentially profound,40 and it was advised to not publish that observation as part of the study. However, alerting the wider oncology community that works with Pt drugs is critical (and others have sought to make researchers aware in other ways).

Key challenges for the field Below, we assess recent trends in the platinum medicine field, and discuss key challenges for metals-in-medicine generally. The relevant pre-existing work is cited in each case, and it is hoped that this will stimulate interest and experimentation in new fields, and entice experts in related fields into entering the waters. We chose four ‘recent trends’ to

ACS Paragon Plus Environment

12

Page 13 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

give a sense of where we see current major developments, and discuss ‘key challenges’ that we believe deserve further investigation (we do not suggest they are novel, just underexplored). There is obviously a skew to these based on our interest in oncology experimental therapeutics and translational science, and we have attempted to discuss the most difficult questions. We are hopeful that these topics will stimulate thought and discussion on the topic, and that they highlight much that lies fallow.

1. Seeing is believing: microdose imaging of clinical and experimental Pt drugs in humans. Image-based insight into drug tumor distribution is a powerful tool during pre-clinical drug development, and for image-guided personalization of therapeutic regimens.43 For example, [18F]paclitaxel (FPAC) has been used for positron emission tomography (PET) imaging to show that uptake levels in breast tumors correlated with clinical response.44 Such tools are under-developed for clinical and investigational Pt-based drugs, and we believe implementation will aid in answering questions related to tumor drug levels, clearance rates, off-target organ residence times, and side effects (perhaps including sites such as the inner ear that present a toxicity challenge). As it stands, the information that exists relating to patient tumor-drug localization has relied on direct measurement of bulk Pt levels in post-surgical tissue from non-small cell lung cancer,45 which demonstrated a correlation between Pt levels and survival for 44 patients, and more recently in a smaller study of muscle-invasive bladder cancer.46 A limitation of these valuable studies is that patients had received Pt chemotherapy over a month prior to tissue resection. While they are single time-point studies, the obvious advantage of directly measuring Pt levels is that

ACS Paragon Plus Environment

13

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 66

the moiety measured is the one that exerts activity, and the challenge is that the proportion of total Pt responsible for cell-killing activity is not known but presumably a very small proportion of total Pt in the cell. Spectroscopies such as X-ray fluorescence spectroscopy (XFS, also known as synchrotron resonance induced X-ray fluorescence, SRIXE), and NanoSIMS (Nano-scale surface ionization mass spectrometry), can provide spatial information on Pt localization, and one imagines a wealth of post-chemotherapy samples are in pathology banks and available that may be amenable to such a study, but the challenge again remains that the limitation is that clinical samples are ex vivo samples from a single time-point.

Radiolabeled Pt complexes have been generated for laboratory studies, and could be used for animal biodistribution47 and time-course studies. Cisplatin has no carbon, and [3H] labeling is not feasible because protons on Pt ammine ligands exchange rapidly in water. Carboplatin has been labeled on the ‘leaving’ cyclobutyldicarboxylate ligand, 48, 49 limiting its utility as one is essentially tracing the non-pharmacologic leaving group rather than the bioactive moiety, particularly over longer time-points. Oxaliplatin’s 1,2cyclohexylamine non-leaving ligand has been [14C] labeled, and this seems the most appropriate Pt radioligand to study for this reason.50 In a similar way, satraplatin and miriplatin contain non-leaving amines useful for [14C] or [3H] labeling, but neither appears to be reported.

An emerging theme in drug development is the concept of Phase 0 studies that precede the usual Phase I dose escalation studies. Phase 0 studies utilize a non-pharmacologic

ACS Paragon Plus Environment

14

Page 15 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

‘microdose’ that allows insight into the human pharmacokinetic profile.51 Ideally, the investigational agent is labeled with [14C] for detection by accelerator mass spectrometry (AMS), or possibly another radioisotope with a long half-life, but less-sensitive mass spectrometry-based detection tools are also utilized. No report currently exists for Pt microdose studies in humans, though it seems achievable for complexes amenable to [14C] labeling (see above), and may be a potential method for comparing a small number of candidate Pt drugs. The development of a pharmacodynamic microdose study that aimed at assessing Pt-DNA adducts in subjects by isolating peripheral blood mononucleocyte cells (PBMCs) as a predictive biomarker was reported in 2013,52 but results have not been disclosed as of this writing.

What is possible for temporal imaging strategies in human subjects with platinum drugs, then? There are four accessible radioisotopes of Pt: 191Pt (t1/2 = 3 days), 193mPt (t1/2 = 4.3 days), 195mPt (t1/2 = 4 days), and 197Pt (t1/2 = 17 hours)53 from which radiolabeled Pt complexes could be generated. By using radiolabeled Pt, the structure of the drug itself is unmodified. All radioisotopes produce gamma emission, and would be amenable to either gamma camera scintigraphy (producing 2D images) or single-photon emission computed tomography (SPECT) imaging (producing 3D images). Studies reporting [191Pt]cisplatin (1985)54 and [193mPt]cisplatin (1973)55 preparation have received followup; [191Pt]cisplatin appears to have been intended partly as a radiotherapeutic pharmacologic form of cisplatin that did not progress into humans,56 and partly for imaging (see below), and the [193mPt]cisplatin was not used for imaging. Multiple reports

ACS Paragon Plus Environment

15

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 66

of preparations of [195mPt]cisplatin exist from 1973 onwards,55, 57-60 and performance assessment showed that the minimum detection limit for a 2 cm tumor was 8 ppm.61

All but one of the handful of imaging studies have utilized [195mPt]cisplatin, and specific examples are shown in Figure 3. The first report with [195mPt]cisplatin displayed a gamma camera image at very low resolution in a 35-year-old woman with metastatic carcinoma of the cervix55 (Fig. 3A), and a similar strategy was independently reported the following year.62 A decade later, Owens et al. produced [191Pt]-labeled cisplatin, carboplatin, and the platinum(IV) clinical candidate, iproplatin, and gamma camera images were used to assess liver and kidney levels in patients with a number of different malignancies, but tumor accumulation itself could not be discerned.63

The first report to utilize imaging for clinical insight came from Wolf and co-workers (1989), who used scintigraphic imaging to assess [195mPt]cisplatin levels in brain tumors of patients receiving intra-venous (i.v.) versus intra-arterial (i.a.)64 administration (intraarterial administration of chemotherapy via the carotid to maximize delivery of drug to brain tumors was of great clinical interest in the 1980s). Scans were collected each minute to create time-activity curves, and tumor signal (ipsilateral) was compared with the non-tumor brain hemisphere (contralateral), revealing lower tumor or brain uptake of cisplatin administered i.v., and 3-10 times increased cisplatin levels when administered i.a. (Fig 3B). Brain tumor could be clearly distinguished because of the very low surrounding brain Pt levels. Areberg et al. imaged [191Pt]cisplatin distribution in 14 patients, and in 5 cases were able to distinguish tumor uptake in patients with a range of

ACS Paragon Plus Environment

16

Page 17 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

malignancies including oral, lung and testis65 (Fig 3C). Critically, tumor location was confirmed by separate computed tomography (CT) scans, and tumor Pt concentrations were derived from quantitated signal and compared against Pt pharmacokinetics. More recently, a study of [195mPt]cisplatin scintigraphy distribution in five healthy volunteers has been reported (along with associated PK), with the highest organ exposure in kidneys (Fig 3D).66

PET allows higher-resolution imaging than SPECT, but also has a number of restrictions associated with it, including the limited number of radioisotopes able to produce positrons upon radioactive decay. While Pt radioisotopes do not appear to be amenable to PET imaging, [11C] and [18F] are the most regularly utilized radioisotopes in PET imaging, though not yet reported for Pt drugs. However, multiple syntheses of [13N]cisplatin have been reported,67-69 the first in 1985. The challenge for clinical imaging is the short half-life of the 13N radioisotope (t1/2 = 10 mins), shorter even than [11C] (t1/2 = 20 min) and [18F] (t1/2 = 1.8 h). Only one report exists using [13N]cisplatin for PET imaging (compared with three reports on its preparation!).70 Ginos et al. assessed biodistribution, blood clearance and stability first in rats, and could not detect displacement of the [13N] ammine ligand from cisplatin in deproteinized blood samples. PET studies were performed in two patients with glioblastoma following both i.v. and i.a. (carotid artery) injection, in conjunction with regular chemotherapy administration, producing brain:tumor concentrations of 30:1, with increasing tumor accumulation over 15 minutes (Fig 3E).70 A.

B.

ACS Paragon Plus Environment

17

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

C.

Page 18 of 66

D.

E.

Figure 3. Examples of images acquired from human subjects treated with radiolabeled cisplatin. A. 1973. 35-year old woman with metastatic carcinoma of the cervix imaged

ACS Paragon Plus Environment

18

Page 19 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

following injection with [195mPt]cisplatin.55 The strong activity arises from high concentrations in the kidneys. This research was originally published in JNM. Lange, R.C., Spencer, R.P., Harder H.C. The Antitumor Agent cis-Pt(NH3)2Cl2: Distribution Studies and Dose Calculations for 193mPt and 195mPt. J Nucl Med. 1973;14:191-195. © by the Society of Nuclear Medicine and Molecular Imaging, Inc. B. Images of a patient with astrocytoma (brain tumor) injected with [195mPt]cisplatin either i.v. (left) or i.a. (right).64 Low brain signal compared to surrounding tissue can be seen at left. The high signal (dark) at right corresponds to the tumor region. C. Static anterior images over the head, neck and thorax region of a patient with metastatic lung cancer immediately after injection with [191Pt]cisplatin, with several tumors arrowed.65 D. Anterior (frontal) view of healthy subject 5 hours following injection of [195mPt]cisplatin.66 E. 1987. Composite 13 minute images of [13N]cisplatin PET scans following i.v. administration, with the drug being infused during the data collection.70 This research was originally published in JNM. Ginos, J.Z., Cooper, A.J.L., Dhawan, V., Lai, J.C.K., Strother, S.C., Alcock, N., Rottenberg, D.A. [13N]Cisplatin PET to Assess Pharmacokinetics of Intra-Arterial Versus Intravenous Chemotherapy for Malignant Brain Tumors. J Nucl Med. 1987;28:18441852. © by the Society of Nuclear Medicine and Molecular Imaging, Inc.

The images collated in Figure 3 demonstrate the evolution of radiolabeled cisplatin over time. Three of the four patient studies (i.e. subjects with tumors) were published in the 1980s, and most of the studies are proofs-of-concept rather than clinical studies. Given that both scintigraphy/SPECT (Pt radioisotope) and PET imaging ([13N]) with cisplatin have been demonstrated as useful—why have they not been explored, both in animal

ACS Paragon Plus Environment

19

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 66

models and humans? There are a litany of clinical challenges for Pt drugs, including understanding the acquisition of resistance, virtually no knowledge at all of tumor drug concentrations, and the opportunities for using imaging for differentiating candidate Pt drugs. Producing a fluorinated analog of a Pt drug such as oxaliplatin (non-leaving amine ligand) is an unexplored possibility, provided the analog retains pharmacologic activity (and therefore relevance). Medical imaging technology has progressed significantly in the past 30 years, and identification of tumor sites can be achieved with [18F]fluorodeoxyglucose (FDG) and MRI/CT to facilitate interpretation of drug localization. Coordination between chemists, biologists and clinicians will be needed for any clinical imaging program, and could ultimately produce a clinical tool for identifying patient populations who would benefit from Pt-containing chemotherapeutic regimens.

2. Time for a new drug Cisplatin and carboplatin are workhorses of the clinic, and oxaliplatin has added a new spectrum of activity. Oxaliplatin was approved in the United States in 2002, and it is stated in the literature that it was first approved for clinical use in 1996 in France71 (reflected in Table 1,72 though we have been unable to identify a primary source for this date). Europe-wide approval for oxaliplatin occurred in 1999.73 Aside from these three approved drugs, a range of Pt complexes have entered clinical trials in the United States and Europe (reviewed extensively in 200774 and 201072), the most notable being the Johnson Matthey products cis,trans,cis-[PtCl2(OH)2(isopropylamine)2], (iproplatin, JM975), cis,trans-[PtCl2(acetato)2(NH3)(cyclohexylamine)]

ACS Paragon Plus Environment

20

Page 21 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

(satraplatin, JM21676), and cis-[PtCl2(NH3)(2-methylpyridine)], (picoplatin, JM473, AMD473, ZD047377), and the multinuclear complex [{trans-PtCl(NH3)2}2-mu-(transPt(NH3)2(H2N(CH2)6-NH2)2)]4+ (BBR3464) developed by the Farrell lab.78 While cisplatin, carboplatin and oxaliplatin are essentially approved worldwide, there are four drugs approved for clinical use in specific nations (Table 1 and Figure 4). Carboplatin was developed with the aim of producing a complex with cisplatin’s activity, but without its toxicity. It is worth considering how the clinical activity of the more recent Pt drugs was identified.

Oxaliplatin (originally called l-OHP) was one of many Pt complexes containing 1,2diaminocyclohexane ligands synthesized based on the promising activity of these complexes.79 It was identified as a highly soluble analog that was active against L1210 murine leukemia cells in mice (the standard for front-line testing at the time) and was active against cisplatin-resistant L1210/DDP cells along with other mouse tumors.80 A study of oxaliplatin’s cytotoxicity against the NCI60 panel of cell lines identified oxaliplatin as highly active against colorectal cancer cell lines,71 and this led to clinical trials with 5-fluorouracil that produced improved responses and led to regulatory approval.71 Despite challenges associated with peripheral neuropathy, oxaliplatin continues to enter clinical trials for efficacy against other agents, including rectal, pancreatic, and stomach cancers.81

Nedaplatin (Figure 4, originally termed 254-S72) was approved for use in Japan in 1995, against a range of malignancies including head & neck cancers.82 It has the same non-

ACS Paragon Plus Environment

21

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 66

leaving cis-ammine ligands as cisplatin and carboplatin, and a glycolato ligand as the leaving group. It was developed by Shionogi Pharmaceutical Company, Japan, based on a desire to develop a drug with lesser side-effects than cisplatin. Nedaplatin was one of a small series of Pt glycolato complexes synthesized with various amine ligands, all of which were shown to be highly soluble.83 Nedaplatin was found to be less toxic than cisplatin, and active against a range of animal xenografts,84 and progressed to clinical trials. While it is generally thought of as a second-line platinum drug,82 a recent Japanese clinical trial comparing nedaplatin plus docetaxel with cisplatin plus docetaxel for advanced or relapsed squamous cell lung cancer found that survival was significantly longer, and side effects lessened, using nedaplatin instead of cisplatin.85 This suggests a greater role in the clinic may lie ahead for nedaplatin, and certainly more trials comparing nedaplatin with first-line drugs such as cisplatin and carboplatin.86

Heptaplatin (Figure 4, originally termed SKI 2053R) was approved in Korea in 1999 for the treatment of gastrointestinal malignancies. 87 The compound was part of a series of over forty [2-substituted-4,5-bis(aminomethyl)-1,3-dioxolane]platinum(II) complexes with bidentate carboxylate leaving groups88—the name heptaplatin evidently arose from the seven-membered ring formed between the diamine non-leaving ligand and Pt. The in vivo activity against L1210 tumors was determined, and the most active complexes were tested against stomach cancer cell lines (for which there are limited treatment options), and found to be superior to cisplatin and carboplatin. Heptaplatin was selected as the lead among other analogs based on its aqueous solubility and stability, low nephrotoxicity, and activity against stomach cancer cell lines.89 Clinical trials in Korea led to approval

ACS Paragon Plus Environment

22

Page 23 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

for use of the agent against advanced gastric cancers, in combination with 5fluorouracil.90

Lobaplatin (Figure 4, originally termed D-19466) was originally developed in Germany by ASTA Pharma, has entered clinical trials in multiple nations,91 and was approved for use in China in 2003. It emerged from a series of Pt analogs containing a 1,2bis(aminomethyl)cyclobutane ligand synthesized at ASTA Pharma, with bidentate lactate as the leaving group.92 It was selected for progression to clinical development based on aqueous solubility and stability, superior cytotoxicity to cisplatin against a number of cancer cell lines including melanoma and hepatoma lines, and an absence of nephrotoxicity in mice at a dose that was efficacious against P388 and L1210 tumors (and was more active than both cisplatin and carboplatin),93 and entered clinical trials. The drug passed through several hands in Germany before being licensed for development in China. It is approved in China for the treatment of metastatic breast and small-cell lung cancer, and acute myelogenous leukemia,94 and has been recommended for combination with paclitaxel as second-line treatment for esophageal cancer.95

Miriplatin (Figure 4) was recently approved in Japan, and is structurally distinct in that it features long alkyl chains on its carboxylate leaving groups (myristates) to confer lipophilicity.96 The complex was specifically created for treatment of hepatocellular carcinoma (HCC). It has been shown that the excipient ethiodized oil (Lipiodol), composed of iodine in fatty acid esters derived from poppy seed, is selectively retained in HCC tumors when administered directly via the hepatic artery (a therapeutic strategy

ACS Paragon Plus Environment

23

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 66

termed transarterial chemoembolization, TACE).97 TACE can act as a tumor drugdelivery strategy, and clinical trials with cisplatin suspended in ethiodized oil have been performed, but suspension of drugs insoluble in the oil presents challenges.98 To address this, Sasaki and co-workers prepared a series of ‘liposoluble’ cyclohexane-1, 2-diamine platinum(II) complexes with long-chain carboxylate (C10 – C24) leaving groups.99 The criteria for lead selection were: ‘suspensibility’ in ethiodized oil; Pt release from the suspension to aqueous saline; and in vivo antitumor activity against L1210 murine leukemia cells, and miriplatin, with myristate (tetradecanoate) leaving groups, was selected for preclinical and clinical trials (summarized in96) and was ultimately approved. Dosage is 70 mg of drug suspended in 3.5 mL of ethiodized oil, administered to the patient once a day through a catheter inserted into the hepatic artery.

H 3N

O

O

O

NH 2 O Pt NH 2 O

Pt H 3N

O

O Heptaplatin

Nedaplatin

O CH3

H2 N

NH 2 O Pt NH 2 O

O

(CH)12

O

(CH)12

Pt O

N H2

CH3 O

Lobaplatin

Miriplatin

ACS Paragon Plus Environment

24

Page 25 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Figure 4. Structures of drugs registered for use in specific countries. In each case the cis non-leaving amines are at the left, and the leaving groups are at the right.

Table 1. Drugs registered for clinical use (structures shown in Figure 1). Drug Cisplatin Carboplatin Oxaliplatin Nedaplatin Lobaplatin Heptaplatin Miriplatin

Registered Worldwide Worldwide Worldwide Japan China Korea Japan

Year 1978 1989 1996 1995 2003 1999 2009

Ref 72 100 72 72 72 101 96

The common requirements in the discovery stage for the newer agents described above were solubility, stability and a reduction in side effects compared with cisplatin. None of these four agents (nedaplatin, heptaplatin, miriplatin, lobaplatin) have been reported to display superior activity to cisplatin/carboplatin, though they have been demonstrated to be comparable. This probably accounts for the fact that these drugs have received little clinical attention in the United States or Europe, and presumably commercial factors are also at play. These newer drugs can in a sense be considered ‘me-too’ drugs102 that were developed based on the known structure-activity rules (SAR) for the ‘big three’ Pt drugs.15 That is, a cis arrangement of two non-leaving amine ligands, and two ‘leaving’ ligands. Oxaliplatin can be excused from the ‘me too’ accusation based on its different scope of activity against colorectal cancer. Miriplatin was specifically developed for a formulation that localized to HCC tumors in the liver, though the point stands that structurally it complies with the SAR of cisplatin. The reality is that in each case these later-generation complexes exist to replace another Pt drug, rather than to introduce a

ACS Paragon Plus Environment

25

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 26 of 66

new spectrum of activity, which is a common motivator for me-too drug development. The challenge is the need for the newer ‘me-too’ Pt agent to demonstrate superiority over the clinical standard-of-care Pt drug, replacing one Pt drug with another – carboplatin succeeded in this, satraplatin did not.

There are three strands for the future development of Pt-based drugs: first, the creation of me-too drugs; second, the continued identification of Pt drug combinations with other agents against specific cancers that provides improved patient benefit; and third, the generation of new Pt compounds that push the boundaries of Pt SAR for testing and identification of novel activity against malignancies poorly responsive to current Pt agents.

We believe the greatest hope for Pt drug development lies in creating and identifying compounds with a new spectrum of activity—be it through phenotypic profiling of cancer cell line sensitivity, or medicinal chemistry driven towards identifying a highly active complex against a malignancy of choice. A major challenge for clinical chemotherapy is drug resistance, and this is the case for most patients enrolled in Phase I/II clinical trials being treated with an experimental drug. It seems unlikely that the solution to clinical resistance to a platinum drug will lie with a new Pt drug.6 This does not mean that there is little prospect for new Pt drugs to enter the clinic, but while testing new Pt compounds in cytotoxicity assays against cisplatin-resistant cell lines may inform on a possible alternative cell-killing mechanism to cisplatin, activity against cisplatin-refractory tumors is unlikely to be the basis of a clinical trial.

ACS Paragon Plus Environment

26

Page 27 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

However, one can envisage a Pt drug with a very similar activity profile to cisplatin that could result in replacement of cisplatin/carboplatin as front line drugs. One scenario could be a Pt drug that does not cause ototoxicity in patients. Ototoxicity in various forms (deafness, tinnitus) is not a dose-limiting toxicity as it does not prevent the patient from receiving the drug, but cisplatin can cause immediate and permanent alterations to a patient’s hearing, and the same is true for carboplatin.103 The replacement of cisplatin with carboplatin in clinical regimens has been careful (why change something that works?), and requires side-by-side clinical trials. For example, in the case of nasopharyngeal cancer, most patients receive cisplatin as the front-line agent, despite the fact that individual trials have shown no difference in treatment efficacy,104 and that carboplatin was better tolerated. Ototoxicity is not avoided though, and while the mechanism has not been definitively determined, a mouse model exists for cisplatininduced ototoxicity that could be a useful basis for counter-screening for Pt drugs that do not induce ototoxicity.105

So what are the questions we should be asking of new agents? New clinical candidates that offer genuinely new activity are needed, including activity against intractable malignancies such as glioblastoma and other brain tumors, melanoma, and pancreatic cancer. Brain tumors are an interesting case in point. In the previous section it was noted that brain signal was extremely low during gamma imaging of radiolabeled cisplatin; this appears to be because clinical platinum drugs are inefficient at crossing the blood-brain barrier (BBB). The brain:plasma ratio for cisplatin and carboplatin is between 1:5 and

ACS Paragon Plus Environment

27

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 28 of 66

1:10,106 whereas peripheral tissues demonstrate Pt levels closer to parity with plasma levels. A Pt development program driven by brain tumor cell accumulation (rather than cell line killing) could result in an agent with an improved activity profile against brain tumors. This is just one example, there are of course many such scenarios where pushing beyond simple phenotypic monolayer cell line killing could open up new avenues for Pt drug discovery.

In that context, what pre-clinical Pt candidates appear to be in the pipeline? Information on Pt candidates progressing towards clinical trials is often limited, but Investigational New Drug approval for a clinical trial (in the US, or equivalent procedures elsewhere) is usually accompanied by a press release. There are also websites indexing clinical trials for various regions that can give insight into open trials with new platinum agents. One challenge is in identifying the structure of investigational agent and in some cases similar structures can be inferred from literature published by founders of a given pharmaceutical company.

The ‘phosphaplatin’ complex, PT-112 (1), is being developed by Phosplatin Therapeutics (USA).107 It is currently listed as being in Phase I clinical trials in the United States to evaluate its safety, tolerability, pharmacokinetics and preliminary clinical effects.108 Several academic publications by the lead author Rathindra Bose (who passed away in 2015) describe the ‘phosphaplatin’ class of complexes (Figure 5). The initial synthesized series including both Pt(II) and Pt(IV) complexes with bidentate pyrophosphate complexes, containing cis ammine (NH3), ethane-1,2-diamine and 1,2-

ACS Paragon Plus Environment

28

Page 29 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

diaminocyclohexane (dach) non-leaving groups.109 The complexes comply with Pt SAR rules, but the pyrophosphato ligand confers high aqueous solubility, and does not hydrolyze over weeks in aqueous solution.110 The dach-containing complex termed ‘pyrodach-2’ demonstrated greater cytotoxicity than cisplatin against cisplatin-resistant cells, in spite of lower cellular accumulation.110 A recent paper further assessed the dach complexes and revealed a lack of in vitro DNA interaction, little role for DNA repair genes in sensitivity, and differential gene expression changes in treated ovarian cancer cells, and the investigators have concluded that an alternative mechanism of action to cisplatin is at play.111 The same paper revealed that the R,R enantiomer of pyrodach-2, ‘RRD2’ was approved as an Investigational New Drug, hence it is the agent designated 1 by Phosplatin Therapeutics. Whether 1 truly acts by a mechanism divorced from DNA binding remains to be proven; the reduced cytotoxicity compared with cisplatin, and the high tolerated doses in mice (>60 mg/kg i.v.)112 is consistent with increased stability just like carboplatin and oxaliplatin, both of which contain bidentate leaving groups. Along with trials in the US (drug to be administered intravenously), Phosplatin Therapeutics have also granted a regional license for development and commercialization of 1 in China and nearby countries.113

ACS Paragon Plus Environment

29

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

O

HO

ONa

H2 N

O

2

O

P

O

H 3N

O

P

O Pt

O

Pt N H

Page 30 of 66

O ONa

H 3N

HO

O O

O

PT-112

Dicycloplatin

O

O

N H N

O Cl

N H

O

Pt N H

O

O

Cl H N N O

O Kowol

Figure 5. Structures of pre-clinical Pt complexes and related structures. 1 (R,R-trans-1,2cyclohexanediamine dihydrogen pyrophosphate)platinum(II)) in development by Phosplatin Therapeutics. Dicycloplatin in development by Sopo-Xingda Pharmaceutical, containing carboplatin (structure highlighted in red, also shown in Figure 1) hydrogenbonded to 1,1-cyclobutane dicarboxylic acid (CBDCA). The ‘Kowol’ complex is not preclinical, but is an exemplar from the literature of a Pt(IV) complex possessing axial ligands with pendant maleimide.

The Pt-containing therapeutic BTP-114114 (2) (structure not disclosed) is in development by Placon Therapeutics (USA, scientific co-founder Stephen Lippard115), which recently spun out from Blend Therapeutics (USA) to focus on Pt drugs.116 2 was recently approved as an Investigational New Drug by the FDA.114 2 has been disclosed to be a pro-drug of cisplatin with a pendant maleimide group, which covalently binds to serum albumin, providing an extended plasma half-life.117 Maleimides react with thiols, and

ACS Paragon Plus Environment

30

Page 31 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

human albumin contains a single thiol at residue 34,9 that has been demonstrated to be accessible to maleimide conjugation.118 Based on the disclosure information, it seems likely that 2 is a Pt(IV) complex with axial ligands containing functionalized maleimide groups. Pt(IV) complexes are thought of as ‘pro-drugs’ that release their axial ligands upon reduction to yield the square planar active Pt(II) complex.8 2 may be similar to a concept reported by Kowol and co-workers in 2013,119 where pendant maleimides were coupled to the axial ligands of Pt(IV) complexes (Figure 5). In the case of the Kowol complexes, the Pt(IV) complexes reacted with albumin rapidly (halftime ~1h)119 and showed tumor regression activity. Lippard and co-workers have published a series of asymmetric Pt(IV) complexes designed to non-covalently associate with albumin via their pendant fatty acids (Figure 6b),120 but no information on an analogous covalent complex have been disclosed. Generally speaking, the albumin-binding pro-drug strategy may in some ways parallel abraxane, the albumin-bound formulation of paclitaxel,121 which was developed to circumvent toxicities associated with paclitaxel and its excipient. Molecules such as Evans Blue are avid albumin binders and used on that basis to assess BBB integrity (or similarly tethered to Pt(IV)).122 Along with prolonged pharmacokinetics,117 2 may also be anticipated to benefit from the leaky tumor effect that can allow for high macromolecule accumulation in tumors.123

ACS Paragon Plus Environment

31

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 32 of 66

Figure 6. Schematic of a likely mechanism of action of a compound similar to 2, based on disclosed description of mechanism.117 A Pt(IV) complex containing cisplatin in the equatorial plane, with a pendant maleimide on an axial ligand (left) conjugates with Cys34 of human albumin in the blood stream. Following prolonged circulation, the Pt(IV) complex is ultimately reduced, yielding cisplain, which can then exert its anticancer activity.

Dicycloplatin is effectively a reformulation of carboplatin, wherein the excipient contains an extra molar equivalent of carboplatin’s leaving group, CBDCA that is believed to hydrogen bond with carboplatin and facilitate its solubilization.124 Developed by Sopo-Xingda Pharmaceutical (China), it has greater solubility and stability in solution than carboplatin, and equivalent activity to carboplatin125 A number of clinical trials have been conducted in China, the most recent reported being a Phase II study comparing paclitaxel plus carboplatin versus paclitaxel plus dicycloplatin in patients with advanced non-small-cell lung cancer,126 with both combinations showing similar response and survival rates, concluding that a Phase III trial of dicycloplatin is warranted to confirm efficacy.

ACS Paragon Plus Environment

32

Page 33 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Along with new small molecules, there has been a significant emphasis on liposomal reformulations of platinum complexes.127, 128 While slightly outside the intended scope of this article, there are some interesting points related to the Pt drug being delivered, and the pro-drugs generated as part of the formulations. The archetypal success story in nanoscale drug formulation is that of pegylated liposomal doxorubicin (marketed as Doxil).129 The weak base doxorubicin is hyper-accumulated within the acidic lumen of the pegylated liposomes, creating a high ‘payload’ per liposome. To date, a physico-chemical property of cisplatin and its counterparts has not been identified that would allow noncovalent trapping at high levels (though there is no reason why a complex that can be acid-trapped cannot be created for liposomal loading). However, several liposomal preparations encapsulating unmodified cisplatin have been reported,130 the most advanced being lipoplatin, a liposomal formulation of encapsulated cisplatin with a loading level of 1:10130 that shows prolonged circulation and diminished toxicities. Lipoplatin has now progressed through Phase I, II and III trials, and it was granted orphan drug status by the EMEA in 2007 for treating pancreatic cancer.131 While clinical trials appear to be ongoing, it is not yet approved for full use in any market. Other liposomal formulations encapsulating unmodified Pt agents are also in pre-clinical development.

Beyond lipoplatin, a panoply of liposomal and other nano-scale drug carriers containing Pt drugs are reported in the literature, some of which are progressing towards clinical trials. These involve a covalent attachment of the Pt complex to the nanocarrier to facilitate high Pt drug payload per nanoparticle. This attachment is conferred either via

ACS Paragon Plus Environment

33

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 34 of 66

the axial ligand of a Pt(IV) pro-drug containing cisplatin, carboplatin or oxaliplatin that is released upon reduction,132 or via coordination of carboxylate leaving ligands (Figure 7). What is intriguing is that, unlike the liposomal formulations of intact Pt drugs, both conjugation strategies involve creating a new chemical entity (NCE) that presumably requires full safety assessment. For either strategy, when the Pt detaches from the nanoparticle it produces a version of the active drug; for Pt(IV) attachment the intact drug is presumably yielded; for carboxylate-tethered Pt(II) drugs, the result is presumably a diaquated complex that is highly reactive133 (this aspect of Pt release does not appear to have been explored in detail).

Tethering strategies have led to Pt nanocarrier formulations that have entered clinical trials. For example, Nanocarrier Co. (Japan) have developed several micellar nanoparticles formed from block co-polymers containing a polyamino acid core that coordinates to Pt(II) via carboxylate ligands, using both cis-diammine (cisplatin analog, termed Nanoplatin, {[Pt(1,2-diaminocyclohexane)]}-loaded polymeric micelles or NC6004) and dach (oxaliplatin analog, termed DACH-Platin, or NC 4016), schematically shown in Figure 7.134 Both forms are currently in clinical trials against a range of malignancies in Japan and the US.135, 136 An alternative leaving group is included in the oxaliplatin analog ProLindac (AP5346) (3),137 where the Pt is loaded onto a hydroxypropylmethacrylamide (HPMA) biocompatible polymer via an O,N chelate (Figure 7). Like DACH-Platin, (3) releases a hydrolyzed form of oxaliplatin. (3) entered Phase I and II clinical trials, but appears to have been discontinued.138

ACS Paragon Plus Environment

34

Page 35 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Figure 7. Schematic demonstrating the chelation of Pt-dach in DACH-Platin and (3). Release from the constructs presumably produces the hydrolyzed form of oxalipatin.

While it is clear that structure-activity rules are being adhered to, for both new small molecules and nanocarrier formulations, there is clearly scope of exploration of new chemotypes and new strategies for improving the activity over cisplatin, and reducing the reactivity and toxicities compared with cisplatin both by modifying pharmacokinetics and targeting the Pt payload to tumors. The literature on drug delivery systems incorporating cisplatin and other active Pt complexes is expanding rapidly and it is likely more will enter clinical trials in the near future.127 Understanding Pt drug mechanism and activity, and utilizing the various drug delivery mechanisms available to create new therapeutic

ACS Paragon Plus Environment

35

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 36 of 66

opportunities clearly leads the way for the next Pt therapeutic to be approved for use in the clinic.

3. A role for Pt in antibody-drug conjugates? The emergence of biologics including antibody-directed therapeutics is changing the chemotherapeutic landscape. A cornerstone of this technology are antibody-drug conjugates (ADCs), where a highly specific antibody is directed to a tumor-specific target, delivering a small molecule cytotoxin.139 There are three components to ADCs. First, a monoclonal antibody directed towards a cancer-specific cell-surface target. Second, to the antibody is tethered a linker that connects with the cytotoxic ‘payload’, that can be cleaved to release the cytotoxin. Third, a highly cytotoxic small molecule is conjugated to the linker. It is critical that the synthetic modification to conjugate the small molecule does not interfere with the activity of the molecule. A number of ADCs are in development. For example, trastuzumab emtansine (marketed as Kadcyla) possesses an antibody that targets HER2-positive cells, and delivers a maytansine-like molecule called DM1, a highly cytotoxic molecule that acts by binding to tubulin.140 A critical need for ADCs is that the conjugated cytotoxin is highly potent, as each ADC delivers a few cytotoxin molecules to the cell at most.139 In fact, molecules usually incorporated into ADCs are often natural product derived molecules targeting microtubules (such as maytansinoid or Monomethyl auristatin) that are too toxic for systemic administration. ADCs allow such toxins to be administered as they are inactive till released where they are localized at the tumor site, improving the therapeutic window for these agents.

ACS Paragon Plus Environment

36

Page 37 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Given the emerging clinical importance of ADCs and other antibody- and immunedirected therapeutic approaches, it is unsurprising that combination chemotherapies with Pt drugs are being explored as part of clinical trials.141 However, to date there is little exploration of the possibility of conjugating Pt drugs to antibodies in order to create ADCs. It is possible that that Pt drugs do not provide adequate cytotoxicity, but the improvement in therapeutic window and reduction in side effects such as ototoxicity may confer an advantage.

The main study published describes a trastuzumab-Pt(II) conjugate to target HER2positive cells.142 In this work, an oxaliplatin-like complex was conjugated to trastuzumab via a dipeptide linker that could be cleaved by the cellular protease cathepsin, rigidified to ensure that the Pt complex cannot reach around and coordinate with the antibody. This is important as it has been shown that cisplatin can coordinate with trastuzumab, though it does not affect trastuzumab’s antibody-binding region.143 A Pt:antibody ratio of 6:1 was reported, and the conjugation of Pt(II) did not affect the binding of trastuzumab to HER2 on breast cancer cells. The trastuzumab-Pt construct resulted in almost four-fold higher cellular Pt levels compared with an equimolar dose of oxaliplatin, though it did not result in increased cytotoxicity. There are other examples of Pt-antibody constructs, for example for utilization in mass cytometry, but these were not created with linkers, nor for therapeutic use.144 Waalboer et al. report the use of Pt(II) complexes as bifunctional ADC linkers (as the linker, not as the active drug) connecting a model fluorophore with trastuzumab as proof-of-concept.145

ACS Paragon Plus Environment

37

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 38 of 66

Away from antibodies, an extensive literature exists on the general concept of targeting cancer cells by appending biomolecules to the axial ligands of Pt(IV) complexes.146 By way of example, axial coordination of peptides that bind to integrins have been reported with the intention of targeting cellular uptake to brain tumor cells.147, 148 While promising, cellular targeting guided by small molecules is less likely to provide the exquisite specificity of antibodies directed against validated oncology targets. Pt ADC-related studies are in their infancy, but given that the therapeutic antibodies, and the chemistry needed to conjugate bioactive small molecules, are already in place, translational studies will answer the question of whether there is a role for Pt here. As with the natural product-derived agents currently used, a survey back through the literature to find the most toxic Pt complexes (without great concern for therapeutic window) may mean a ‘new’ Pt chemotype will become a clinical candidate.

4. Chemical biology for cellular fate and mechanisms of action of cisplatin. The verdant field of chemical biology is underpinned by the use of chemical tools to gain insight into biological processes, including understanding of the mechanism of action of small molecule therapeutics. For example, target identification for a drug can be achieved with modified molecules containing a ‘click-able’ pendant group that can be used for proteomic identification of protein targets of a small molecule, or fluorescent labeling to monitor cellular fate. For Pt drugs, the development of chemical biology is occurring at a time when new questions are being asked of the cellular targets of Pt drugs.149 For

ACS Paragon Plus Environment

38

Page 39 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

example, it is clear that Pt drugs interact with a range of protein targets, and understanding their mechanistic relevance (or lack of it) will require significant effort.

Tracking Pt drugs in cells, and their interaction with biomolecules, is not straightforward. As mentioned earlier, few labeling strategies are available for native Pt drugs (i.e., without modification of their chemical structure). This has led to the development of Pt complexes with fluorophores, usually tethered to an amine ligand.150 Examples include complexes conjugated to the fluorophores carboxyfluorescein diacetate (CFDA),151 N-(7nitro-2,1,3-benzoxadiazol-4-yl)ethane-1,2-diamine (NBD),152 and BODIPY.153, 154 These complexes have been used to track the localization of the complexes in a range of cell lines. An obvious limitation to adding a fluorophore to a small inorganic complex is that it may significantly alter the physicochemical properties and biological activity of the Pt complex, and is limited by the assumption that the Pt-fluorophore connection is intact (otherwise it is free fluorophore being imaged).154

To address these limitations, and to allow ‘label-free’ study of Pt drug fate, a number of alternative strategies have been pursued. For example, New and colleagues reported a fluorescent sensor that was used to ‘post-label’ monofunctional Pt species in fixed cells to image its localization.155 Pt complexes have been reported that incorporate minimal structural changes to add a reactive handle for direct post-labeling of the complex. Bierbach and colleagues reported a Pt-acridine with a pendant azide group, and following cell treatment and fixation ‘click’ chemistry was used to append a alkyne-functionalized Alexafluor 488 dye.156

ACS Paragon Plus Environment

39

Cl

N N3

Pt NH 3

Cl

Biological sample

Page 40 of 66

N N3

Pt NH 3

Reporter

Picazoplatin (Picoplatin)

H2 N

Cl Pt Cl

N3 N H2

Target

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Target

Journal of Medicinal Chemistry

N N

Pt NH 3

N N

Reporter

Figure 8. Scheme showing the structure of picazoplatin (with the structure of the clinical candidate picoplatin shown in red), and its reaction with biomolecules. Subsequently ‘click’ chemistry can be used to conjugate an alkyne-functionalized reporter of choice (blue) to the bound picazoplatin via its pendant azide. A Pt complex containing a 2-azido1,3-propanediamine non-leaving bidentate ligand is also shown at bottom left.

In this context, a series of studies by the DeRose group have reported modified Pt drugs with functionalizations that allow chemical biological post-labeling experiments to be performed, such as protein target identification, DNA and RNA binding studies, and incell detection.157 By incorporating an azide or alkyne handle for Click chemistry, relatively small changes to the host Pt complex can be made. The initial study reported an analog of picoplatin with an azide group appended to the 2-methylpyridine ligand

ACS Paragon Plus Environment

40

Page 41 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

(termed picazoplatin, Figure 8), that could be post-labeled with a fluorophore after reaction with DNA or RNA in vitro.158 RNA extracted from yeast cells (S. cerevisiae) treated with picazoplatin were post-labeled with Alexa Fluor 488 to demonstrate that tRNA is a cellular substrate for Pt drugs.159 A Pt complex containing a 2-azido-1,3propanediamine non-leaving bidentate ligand (Figure 8) was shown by post-labeling to bind with bovine serum albumin as a model target protein.160 Confocal images of a similar Pt complex post-labeled in HeLa cells demonstrated the potential for cell-based studies.161 The chemical biology tools now exist to interrogate the cellular targets of Pt drugs, both nucleic acids and protein, and to extend into detailed in vivo studies.

Conclusions Platinum drugs play a critical role in the clinic, and will continue to do so for many years to come. While Pt drugs have been extensively studied, new fields of biology pave the way for new discoveries and understanding of Pt drugs. The development of new Pt drugs that enter clinical trials is a realistic and achievable goal, but requires boldness and sensitivity to the challenges that current Pt drugs can present to patients in the clinic: efficacy that is tempered by side-effects.

ACS Paragon Plus Environment

41

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 42 of 66

Corresponding Author *Matthew Hall, NIH/NCATS, 9800 Medical Center Drive, Rockville, Maryland, 20850 Telephone: (301) 217-5727; Fax: (301) 217-5736; E-mail: [email protected]

Biographies: Dorian Cheff received her Bachelor of Science degree in Neuroscience from the University of Michigan in 2015. She is now at the National Center for Advancing Translational Science, NIH as a post-baccalaureate Intramural Research Training Award (IRTA) Fellow working on assay development and high-throughput drug screening for potential oncology-focused small molecule therapeutics. Her current research interests include efforts to screen Pt compound libraries.

Matthew Hall received his B.Sc. (Hons) in Science and his Ph.D. in Chemistry from the University of Sydney, Australia, under Professor T. W. Hambley, working on understanding the mechanism of action of platinum(IV) complexes. Following a 1-year postdoctoral fellowship with Professor Val C. Culotta at the Johns Hopkins School of Public Health, he joined the Laboratory of Cell Biology, National Cancer Institute (NCI), National Institutes of Health (NIH), under Dr. Michael Gottesman, studying (in part) the mechanisms of cisplatin resistance and multidrug resistance. He joined NCATS in 2015, where his group engages in assay development and high-throughput screening for earlystage small molecule probe discovery, with an emphasis on challenges in oncology drug discovery, including challenges associated with Pt drug research.

ACS Paragon Plus Environment

42

Page 43 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Abbreviations Used ADC, antibody-drug conjugate; AMS, accelerator mass spectrometry; BBB, blood-brain barrier; CBDCA, 1,1-cyclobutanedicarboxylic acid; CFDA, carboxyfluorescein diacetate; CT, computed tomography; dach, 1,2-diaminocyclohexane; FPAC, [18F]paclitaxel; HCC, hepatocellular carcinoma; HPMA, hydroxyprpylmethacrylamide; I.A, intra-arterial; I.V., intravenous; NanoSIMS, nano-scale surface ionization mass spectrometry; NBD, N-(7nitro-2,1,3-benzoxadiazol-4-yl)ethane-1,1-diamine; NCE, new chemical entity; PBMC, peripheral blood mononucleocyte; PET, positron emission tomography; SAR, structureactivity relationships; SPECT, single photon emission computed tomography; TACE, trans-arterial chemoembolization; XFS/SRIXE, x-ray fluorescence spetroscopy

Acknowledgements The authors wish to thank Kyle Brimacombe for his work in designing our figures. This research was supported [in part] by the Intramural Research Program of the NIH, National Center for Advancing Translational Sciences.

References 1.

Shakespeare, W. Cymbeline; 1623.

2.

Hoeschele, J. D. Serendipity and the Prepared Mind: The Discovery of Cisplatin.

F&M Scientist 2015, 3, 54-68. 3.

Kauffman, G. B.; Pentimalli, R.; Doldi, S.; Hall, M. D. Michele Peyrone (1813-

1883), Discoverer of Cisplatin. Platinum Met. Rev. 2010, 54, 250-256.

ACS Paragon Plus Environment

43

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

4.

Page 44 of 66

Kauffman, G. B. Alfred Werner, Founder of Coordination Chemistry; 1 ed.;

Springer-Verlag Berlin Heidelberg: 1966. 5.

Callaghan, R. Providing a Molecular Mechanism for P-Glycoprotein; Why Would

I Bother? Biochem. Soc. Trans. 2015, 43, 995-1002. 6.

Shen, D. W.; Pouliot, L. M.; Hall, M. D.; Gottesman, M. M. Cisplatin Resistance:

A Cellular Self-Defense Mechanism Resulting from Multiple Epigenetic and Genetic Changes. Pharmacol. Rev. 2012, 64, 706-721. 7.

Hall, M. D.; Okabe, M.; Shen, D. W.; Liang, X. J.; Gottesman, M. M. The Role of

Cellular Accumulation in Determining Sensitivity to Platinum-Based Chemotherapy. Annu. Rev. Pharmacol. Toxicol. 2008, 48, 495-535. 8.

Hall, M. D.; Mellor, H. R.; Callaghan, R.; Hambley, T. W. Basis for Design and

Development of Platinum(IV) Anticancer Complexes. J. Med. Chem. 2007, 50, 34033411. 9.

Hall, M. D.; Hambley, T. W. Platinum(IV) Antitumour Compounds: Their

Bioinorganic Chemistry. Coord. Chem. Rev. 2002, 232, 49-67. 10.

Fojo, T. Commentary: Novel Therapies for Cancer: Why Dirty Might Be Better.

Oncologist 2008, 13, 277-283. 11.

Taylor, M.; Filby, A. Health Impact Analysis of Cisplatin, Carboplatin, and

Oxaliplatin. Johnson Matthey Technol. Rev. 2017, 61, 32-39. 12.

Chabner, B. A.; Roberts, T. G. Chemotherapy and the War on Cancer. Nat. Rev.

Cancer 2005, 5, 65-72.

ACS Paragon Plus Environment

44

Page 45 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

13.

Cleare, M. J.; Hoeschele, J. D. Studies on the Antitumor Activity of Group VIII

Transition Metal Complexes. Part I. Platinum (II) Complexes. Bioinorg. Chem. 1973, 2, 187-210. 14.

Silverman, A. P.; Bu, W.; Cohen, S. M.; Lippard, S. J. 2.4-Å Crystal Structure of

the Asymmetric Platinum Complex [Pt(Ammine)(Cyclohexylamine)]2+ Bound to a Dodecamer DNA Duplex. J. Biol. Chem. 2002, 277, 49743-49749. 15.

Hambley, T. W. The Influence of Structure on the Activity and Toxicity of Pt

Anti-Cancer Drugs. Coord. Chem. Rev. 1997, 166, 181-223. 16.

Van Beusichem, M.; Farrell, N. Activation of the trans Geometry in Platinum

Antitumor Complexes. Synthesis, Characterization, and Biological Activity of Complexes with the Planar Ligands Pyridine, N-Methylimidazole, Thiazole, and Quinoline. Crystal and Molecular Structure of trans-Dichlorobis(Thiazole)Platinum(II). Inorg. Chem. 1992, 31, 634-639. 17.

Hearn, J. M.; Romero-Canelon, I.; Munro, A. F.; Fu, Y.; Pizarro, A. M.; Garnett,

M. J.; Mcdermott, U.; Carragher, N. O.; Sadler, P. J. Potent Organo-Osmium Compound Shifts Metabolism in Epithelial Ovarian Cancer Cells. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, E3800-3805. 18.

Google. Google Scholar. http://scholar.google.com/ (August 11, 2016).

19.

Sancton, T.; Le Quesne, N. From Despair to Glory. Time International July 19,

1999. 20.

Armstrong, L. It's Not About the Bike: My Journey Back to Life; The Berkley

Publishing Group: New York, New York, 2000.

ACS Paragon Plus Environment

45

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

21.

Page 46 of 66

Schnell, F. M. Chemotherapy-Induced Nausea and Vomiting: The Importance of

Acute Antiemetic Control. Oncologist 2003, 8, 187-198. 22.

Karasawa, T.; Steyger, P. S. An Integrated View of Cisplatin-Induced

Nephrotoxicity and Ototoxicity. Toxicol. Lett. 2015, 237, 219-227. 23.

The Discovery, Use and Impact of Platinum Salts as Chemotherapy Agents for

Cancer; In Wellcome Witnesses to Twentieth Century Medicine Seminars; Christie DA, T. E., Ed; The Wellcome Trust Centre for the History of Medicine at UCL; 2006, 30 24.

Carter, S. K.; Cleare, M. J.; Connors, T. A.; Conran, P. B.; Dienstbier, Z.; Ellerby,

R. A.; Harder, M. C.; Hill, J. M.; Hindy, I.; Krakoff, I.; Roberts, J. J.; Robins, A. B.; Talley, R. W.; Thomson, A. J.; Wallace, H. J.; Williams, R. J.; Wiltshaw, E. Platinum Coordination Complexes in Cancer Chemotherapy; Springer-Verlag New York: Berlin, Germany, 1974. 25.

Hill, J. M.; Loeb, E.; Maclellan, A. S.; Hill, N. O.; Khan, A.; Kogler, J. Further

Clinical Experience with cis-Platinum(II) Diamminedichloride; In Platinum Coordination Complexes in Cancer Chemotherapy, Connors, T. A., Roberts, J.J., Ed. Springer-Verlag New York: Berlin, 1974; pp 145-152. 26.

Calvert, A. H.; Harland, S. J.; Newell, D. R.; Siddik, Z. H.; Jones, A. C.;

Mcelwain, T. J.; Raju, S.; Wiltshaw, E.; Smith, I. E.; Baker, J. M.; Peckham, M. J.; Harrap, K. R. Early Clinical Studies with cis-Diammine-1,1-Cyclobutane Dicarboxylate Platinum II. Cancer Chemother. Pharmacol. 1982, 9, 140-147. 27.

Goodfield, J. Cancer under Siege; Hutchinson: London, UK, 1975.

28.

Cisplatin: Chemistry and Biochemistry of a Leading Anticancer Drug; Verlag

Helvetica Chimica Aeta and WILEY-VCH: Zürich, Switzerland, 1999.

ACS Paragon Plus Environment

46

Page 47 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

29.

Twitter. https://twitter.com/ (August 11, 2016).

30.

Descoeur, J.; Pereira, V.; Pizzoccaro, A.; Francois, A.; Ling, B.; Maffre, V.;

Couette, B.; Busserolles, J.; Courteix, C.; Noel, J.; Lazdunski, M.; Eschalier, A.; Authier, N.; Bourinet, E. Oxaliplatin-Induced Cold Hypersensitivity Is Due to Remodelling of Ion Channel Expression in Nociceptors. EMBO Mol. Med. 2011, 3, 266-278. 31.

Schierl, R.; Rohrer, B.; Hohnloser, J. Long-Term Platinum Excretion in Patients

Treated with Cisplatin. Cancer Chemother. Pharmacol. 1995, 36, 75-78. 32.

Theon, A. P.; Pascoe, J. R.; Carlson, G. P.; Krag, D. N. Intratumoral

Chemotherapy with Cisplatin in Oily Emulsion in Horses. J. Am. Vet. Med. Assoc. 1993, 202, 261-267. 33.

Theon, A. P.; Wilson, W. D.; Magdesian, K. G.; Pusterla, N.; Snyder, J. R.;

Galuppo, L. D. Long-Term Outcome Associated with Intratumoral Chemotherapy with Cisplatin for Cutaneous Tumors in Equidae: 573 Cases (1995-2004). J. Am. Vet. Med. Assoc. 2007, 230, 1506-1513. 34.

Knapp, D. W.; Richardson, R. C.; Denicola, D. B.; Long, G. G.; Blevins, W. E.

Cisplatin Toxicity in Cats. J. Vet. Intern. Med. 1987, 1, 29-35. 35.

Bell, J.; Brady, M. F.; Young, R. C.; Lage, J.; Walker, J. L.; Look, K. Y.; Rose,

G. S.; Spirtos, N. M. Randomized Phase III Trial of Three Versus Six Cycles of Adjuvant Carboplatin and Paclitaxel in Early Stage Epithelial Ovarian Carcinoma: A Gynecologic Oncology Group Study. Gynecol. Oncol. 2006, 102, 432-439. 36.

Vasey, P. A.; Jayson, G. C.; Gordon, A.; Gabra, H.; Coleman, R.; Atkinson, R.;

Parkin, D.; Paul, J.; Hay, A.; Kaye, S. B. Phase III Randomized Trial of Docetaxel-

ACS Paragon Plus Environment

47

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 48 of 66

Carboplatin Versus Paclitaxel-Carboplatin as First-Line Chemotherapy for Ovarian Carcinoma. J. Natl. Cancer Inst. 2004, 96, 1682-1691. 37.

Holmes, D. Ovarian Cancer: Beyond Resistance. Nature 2015, 527, S217.

38.

Holmes, D. The Problem with Platinum. Nature 2015, 527, S218-219.

39.

Colvis, C. M.; Austin, C. P. Innovation in Therapeutics Development at the

NCATS. Neuropsychopharmacology 2014, 39, 230-232. 40.

Hall, M. D.; Telma, K. A.; Chang, K. E.; Lee, T. D.; Madigan, J. P.; Lloyd, J. R.;

Goldlust, I. S.; Hoeschele, J. D.; Gottesman, M. M. Say No to DMSO: Dimethylsulfoxide Inactivates Cisplatin, Carboplatin, and Other Platinum Complexes. Cancer Res. 2014, 74, 3913-3922. 41.

Lowe, D. DMSO Will Ruin Your Platinum Drugs. Take Heed.

http://blogs.sciencemag.org/pipeline/archives/2014/05/12/dmso_will_ruin_your_platinum _drugs_take_heed (July 29, 2016) 42.

Kerrison, S. J. S.; Sadler, P. J. Solvolysis of cis-[Pt(Nh3)2Cl2] in Dimethyl

Sulphoxide and Reactions of Glycine with [PtCl3(Me2SO)]– as Probed by 195pt Nuclear Magnetic Resonance Shifts and 195Pt–15n Coupling Constants. Chem. Commun. (Cambridge, U. K.) 1977, 861-863. 43.

De Vries, E. G.; De Jong, S.; Gietema, J. A. Molecular Imaging as a Tool for

Drug Development and Trial Design. J. Clin. Oncol. 2015, 33, 2585-2587. 44.

Kurdziel, K. A.; Kalen, J. D.; Hirsch, J. I.; Wilson, J. D.; Agarwal, R.; Barrett, D.;

Bear, H. D.; Mccumiskey, J. F. Imaging Multidrug Resistance with 4[18F]Fluoropaclitaxel. Nucl. Med. Biol. 2007, 34, 823-831.

ACS Paragon Plus Environment

48

Page 49 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

45.

Kim, E. S.; Lee, J. J.; He, G.; Chow, C. W.; Fujimoto, J.; Kalhor, N.; Swisher, S.

G.; Wistuba, Ii; Stewart, D. J.; Siddik, Z. H. Tissue Platinum Concentration and Tumor Response in Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2012, 30, 3345-3352. 46.

Guancial, E. A.; Kilari, D.; Xiao, G.-Q.; Abu-Farsakh, S. H.; Baran, A.; Messing,

E. M.; Kim, E. S. Platinum Concentration and Pathologic Response to Cisplatin-Based Neoadjuvant Chemotherapy in Muscle-Invasive Bladder Cancer. PLoS ONE 2016, 11, e0155503. 47.

Hoeschele, J. D.; Butler, T. A.; Roberts, J. A. Correlations of Physico-Chemical

and Biological Properties with in Vivo Biodistribution Data for Platinum-195m-Labeled Chloroammineplatinum(II) Complexes. ACS Symp. Ser. 1980, 140, 181-208. 48.

Shen, D. W.; Liang, X. J.; Gawinowicz, M. A.; Gottesman, M. M. Identification

of Cytoskeletal [14C]Carboplatin-Binding Proteins Reveals Reduced Expression and Disorganization of Actin and Filamin in Cisplatin-Resistant Cell Lines. Mol. Pharmacol. 2004, 66, 789-793. 49.

Hah, S. S.; Stivers, K. M.; De Vere White, R. W.; Henderson, P. T. Kinetics of

Carboplatin-DNA Binding in Genomic DNA and Bladder Cancer Cells as Determined by Accelerator Mass Spectrometry. Chem. Res. Toxicol. 2006, 19, 622-626. 50.

Jong, N. N.; Nakanishi, T.; Liu, J. J.; Tamai, I.; Mckeage, M. J. Oxaliplatin

Transport Mediated by Organic Cation/Carnitine Transporters OCTN1 and OCTN2 in Overexpressing Human Embryonic Kidney 293 Cells and Rat Dorsal Root Ganglion Neurons. J. Pharmacol. Exp. Ther. 2011, 338, 537-547. 51.

Rani, P. U.; Naidu, M. U. Phase 0 - Microdosing Strategy in Clinical Trials.

Indian J. Pharmacol. 2008, 40, 240-242.

ACS Paragon Plus Environment

49

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

52.

Page 50 of 66

Cimino, G. D.; Pan, C. X.; Henderson, P. T. Personalized Medicine for Targeted

and Platinum-Based Chemotherapy of Lung and Bladder Cancer. Bioanalysis 2013, 5, 369-391. 53.

Dowell, J. A.; Sancho, A. R.; Anand, D.; Wolf, W. Noninvasive Measurements

for Studying the Tumoral Pharmacokinetics of Platinum Anticancer Drugs in Solid Tumors. Adv. Drug Delivery Rev. 2000, 41, 111-126. 54.

Baer, J.; Harrison, R.; Mcauliffe, C. A.; Zaki, A.; Sharma, H. L.; Smith, A. G.

Microscale Syntheses of Anti-Tumour Platinum Compounds Labelled with 191Pt. Int. J. Appl. Radiat. Isot. 1985, 36, 181-184. 55.

Lange, R. C.; Spencer, R. P.; Harder, H. C. The Antitumor Agent cis-

Pt(NH3)2Cl2: Distribution Studies and Dose Calculations for 193m Pt and 195m Pt. J. Nucl. Med. 1973, 14, 191-195. 56.

Areberg, J.; Wennerberg, J.; Johnsson, A.; Norrgren, K.; Mattsson, S. Antitumor

Effect of Radioactive Cisplatin (191Pt) on Nude Mice. Int. J. Radiat. Oncol., Biol., Phys. 2001, 49, 827-832. 57.

Smith, S. V.; Cassidy, D.; Di Bartolo, N. Rapid and Reliable Synthesis of

195mPt-Cisplatin and 195mPt-Carboplatin for Use in Prognosis of Cancer. J. Labelled Compd. Radiopharm. 2001, 44, S756-S758. 58.

Zeevaart, J. R.; Wagener, J.; Marjanovic-Painter, B.; Sathekge, M.; Soni, N.;

Zinn, C.; Perkins, G.; Smith, S. V. Production of High Specific Activity (195m) PtCisplatinum at South African Nuclear Energy Corporation for Phase 0 Clinical Trials in Healthy Individual Subjects. J. Labelled Compd. Radiopharm. 2013, 56, 495-503.

ACS Paragon Plus Environment

50

Page 51 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

59.

Anand, D.; Wolf, W. A New, Semi-Automated System for the Micro-Scale

Synthesis of [195mPt]Cisplatin Suitable for Clinical Studies. Int. J. Rad. Appl. Instrum. A 1992, 43, 809-814. 60.

Aalbersberg, E.; De Wit - Van Der Veen, L.; Zwaagstra, O.; Codee - Van Der

Schilden, K.; Vegt, E.; Vogel, W. Preclinical Imaging Characteristics of 195m-Platinum Spect. J. Nucl. Med. 2014, 55, 1382. 61.

Buckley, S. E.; Ali, P. A.; Evans, C. J.; El-Sharkawi, A. M. Gamma Camera

Scintigraphy of Tumours Using (195m)Pt-Cisplatin. Phys. Med. Biol. 2006, 51, 13251332. 62.

Smith, H. S.; Taylor, D. M. Distribution and Retention of the Antitumor Agent

195mPt-cis-Dichlorodiammine Platinum (II) in Man. J. Nucl. Med. 1974, 15, 349-351. 63.

Owens, S. E.; Thatcher, N.; Sharma, H.; Adam, N.; Harrison, R.; Smith, A.; Zaki,

A.; Baer, J. C.; Mcauliffe, C. A.; Crowther, D.; Fox, B. W. In Vivo Distribution Studies of Radioactively Labelled Platinum Complexes; cis-Dichlorodiammine Platinum(II), cistrans-Dichlorodihydroxy-bis-(Isopropylamine) Platinum(IV), cis-Dichloro-bisCyclopropylamine Platinum(II), and cis-Diammine 1,1-Cyclobutanedicarboxylate Platinum(II) in Patients with Malignant Disease, Using a Gamma Camera. Cancer Chemother. Pharmacol. 1985, 14, 253-257. 64.

Shani, J.; Bertram, J.; Russell, C.; Dahalan, R.; Chen, D. C.; Parti, R.; Ahmadi, J.;

Kempf, R. A.; Kawada, T. K.; Muggia, F. M.; Wolf, W. Noninvasive Monitoring of Drug Biodistribution and Metabolism: Studies with Intraarterial Pt-195m-Cisplatin in Humans. Cancer Res. 1989, 49, 1877-1881.

ACS Paragon Plus Environment

51

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

65.

Page 52 of 66

Areberg, J.; Bjorkman, S.; Einarsson, L.; Frankenberg, B.; Lundqvist, H.;

Mattsson, S.; Norrgren, K.; Scheike, O.; Wallin, R. Gamma Camera Imaging of Platinum in Tumours and Tissues of Patients after Administration of 191Pt-Cisplatin. Acta Oncol. 1999, 38, 221-228. 66.

Sathekge, M.; Wagener, J.; Smith, S. V.; Soni, N.; Marjanovic-Painter, B.; Zinn,

C.; Van De Wiele, C.; D'asseler, Y.; Perkins, G.; Zeevaart, J. R. Biodistribution and Dosimetry of 195mPt-Cisplatin in Normal Volunteers. Imaging Agent for Single Photon Emission Computed Tomography. Nuklearmedizin 2013, 52, 222-227. 67.

Holschbach, M.; Hamkens, W.; Steinbach, A.; Hamacher, K.; Stöcklin, G.

[13N]Cisplatin: A Fast and Efficient On-Line Synthesis Using a Solid State Support. Appl. Radiat. Isot. 1997, 48, 739-744. 68.

De Spiegeleer, B.; Slegers, G.; Vandecasteele, C.; Van Den Bossche, W.;

Schelstraete, K.; Claeys, A.; De Moerloose, P. Microscale Synthesis of Nitrogen-13Labeled Cisplatin. J. Nucl. Med. 1986, 27, 399-403. 69.

Haber, M. T.; Cooper, A. J. L.; Rosenspire, K. C.; Ginos, J. Z.; Rottenberg, D. A.

Synthesis of [13N]Cisplatin. J. Labelled Compd. Radiopharm. 1985, 22, 509-516. 70.

Ginos, J. Z.; Cooper, A. J.; Dhawan, V.; Lai, J. C.; Strother, S. C.; Alcock, N.;

Rottenberg, D. A. [13N]Cisplatin PET to Assess Pharmacokinetics of Intra-Arterial Versus Intravenous Chemotherapy for Malignant Brain Tumors. J. Nucl. Med. 1987, 28, 1844-1852. 71.

Graham, J.; Muhsin, M.; Kirkpatrick, P. Oxaliplatin. Nat. Rev. Drug Discovery

2004, 3, 11-12.

ACS Paragon Plus Environment

52

Page 53 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

72.

Wheate, N. J.; Walker, S.; Craig, G. E.; Oun, R. The Status of Platinum

Anticancer Drugs in the Clinic and in Clinical Trials. Dalton Trans. 2010, 39, 8113-8127. 73.

Bécouarn, Y.; Agostini, C.; Trufflandier, N.; Boulanger, V. Oxaliplatin: Available

Data in Non-Colorectal Gastrointestinal Malignancies. Crit. Rev. Oncol. Hematol. 2001, 40, 265-272. 74.

Boulikas, T.; Pantos, A.; Bellis, E.; Christofis, P. Designing Platinum Compounds

in Cancer: Structures and Mechanisms. Cancer Ther. 2007, 5, 537-583. 75.

Clavel, M.; Monfardini, S.; Gundersen, S.; Kaye, S.; Siegenthaler, P.; Renard, J.;

Van Glabbeke, M.; Pinedo, H. M. Phase II Study of Iproplatin (CHIP, JM-9) in Advanced Testicular Cancers Progressing after Prior Chemotherapy. Eur. J. Cancer Clin. Oncol. 1988, 24, 1345-1348. 76.

Bhargava, A.; Vaishampayan, U. N. Satraplatin: Leading the New Generation of

Oral Platinum Agents. Expert Opin. Invest. Drugs 2009, 18, 1787-1797. 77.

Kelland, L. R.; Sharp, S. Y.; O’Neill, C. F.; Raynaud, F. I.; Beale, P. J.; Judson, I.

R. Mini-Review: Discovery and Development of Platinum Complexes Designed to Circumvent Cisplatin Resistance. J. Inorg. Biochem. 1999, 77, 111-115. 78.

Manzotti, C.; Pratesi, G.; Menta, E.; Di Domenico, R.; Cavalletti, E.; Fiebig, H.

H.; Kelland, L. R.; Farrell, N.; Polizzi, D.; Supino, R.; Pezzoni, G.; Zunino, F. BBR 3464: A Novel Triplatinum Complex, Exhibiting a Preclinical Profile of Antitumor Efficacy Different from Cisplatin. Clin. Cancer Res. 2000, 6, 2626-2634. 79.

Misset, J. L.; Bleiberg, H.; Sutherland, W.; Bekradda, M.; Cvitkovic, E.

Oxaliplatin Clinical Activity: A Review. Crit. Rev. Oncol. Hematol. 2000, 35, 75-93.

ACS Paragon Plus Environment

53

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

80.

Page 54 of 66

Tashiro, T.; Kawada, Y.; Sakurai, Y.; Kidani, Y. Antitumor Activity of a New

Platinum Complex, Oxalato (trans-L-1,2-Diaminocyclohexane)Platinum (II): New Experimental Data. Biomed. Pharmacother. 1989, 43, 251-260. 81.

National Cancer Institute. Clinical Trials Information for Patients and Caregivers.

http://www.cancer.gov/about-cancer/treatment/clinical-trials (August 11, 2016). 82.

Shimada, M.; Itamochi, H.; Kigawa, J. Nedaplatin: A Cisplatin Derivative in

Cancer Chemotherapy. Cancer Manage. Res. 2013, 5, 67-76. 83.

Totani, T.; Aono, K.; Komura, M.; Adachi, Y. Synthesis of (Glycolato-

O,O')Diammineplatinum(II) and Its Related Complexes. Chem. Lett. 1986, 15, 429-432. 84.

Shiratori, O. Anti-Tumor Activity of 254-S, a Platinum Complex, in Rodents.

Recent Adv. Chemother., Proc. Int. Congr. Chemother., 14th 1985, 635-636. 85.

Shukuya, T.; Yamanaka, T.; Seto, T.; Daga, H.; Goto, K.; Saka, H.; Sugawara, S.;

Takahashi, T.; Yokota, S.; Kaneda, H.; Kawaguchi, T.; Nagase, S.; Oguri, T.; Iwamoto, Y.; Nishimura, T.; Hattori, Y.; Nakagawa, K.; Nakanishi, Y.; Yamamoto, N. Nedaplatin Plus Docetaxel Versus Cisplatin Plus Docetaxel for Advanced or Relapsed Squamous Cell Carcinoma of the Lung (WJOG5208L): A Randomised, Open-Label, Phase 3 Trial. Lancet Oncol. 2015, 16, 1630-1638. 86.

Wu, Y.; Spicer, J. Nedaplatin: A New Platinum for Squamous Lung Cancer?

Lancet Oncol. 2015, 16, 1573-1574. 87.

Kim, D. K.; Ahn, J. S.; Ryu, G.; Kim, K. H.; Park, C. W.; Kim, M. S.; Chung, M.

H.; Shin, S. G.; Suh, Y. H.; Kim, Y. S. General Pharmacology of cis-Malonato[4R,5R)4,5-bis-(Aminomethyl)-2-Isopropyl-1,3- Dioxolane]Platinum(II). Drug Res. (Stuttgart, Ger.) 1994, 44, 1080-1088.

ACS Paragon Plus Environment

54

Page 55 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

88.

Kim, D.-K.; Kim, G.; Gam, J.; Cho, Y.-B.; Kim, H.-T.; Tai, J.-H.; Kim, K. H.;

Hong, W.-S.; Park, J.-G. Synthesis and Antitumor Activity of a Series of [2-Substituted4,5-bis(Aminomethyl)-1,3-Dioxolane]Platinum(II) Complexes. J. Med. Chem. 1994, 37, 1471-1485. 89.

Kim, D.-K.; Kim, H.-T.; Cho, Y.-B.; Tai, J. H.; Ahn, J. S.; Kim, T.-S.; Kim, K.

H.; Hong, W.-S. Antitumor Activity of cis-Malonato[(4R,5R)-4,5-bis(Aminomethy1)-2Isopropyl-1,3-Dioxolanelplatinum(II), a New Platinum Analogue, as an Anticancer Agent. Cancer Chemother. Pharmacol. 1995, 35, 441-445. 90.

Min, Y. J.; Bang, S.-J.; Shin, J. W.; Kim, D. H.; Park, J. H.; Kim, G. Y.; Ko, B.

K.; Choi, D. H.; Cho, H. R. Combination Chemotherapy with 5-Fluorouracil and Heptaplatin as First-Line Treatment in Patients with Advanced Gastric Cancer. J. Korean Med. Sci. 2004, 19, 369-373. 91.

Mckeage, M. J. Lobaplatin: A New Antitumour Platinum Drug. Expert Opin.

Invest. Drugs 2001, 10, 119-128. 92.

Schumacher, W.; Respondek, J.; J, E.; Hitzel-Zerrahn, F.; Voegeli, R.; Pohl, J.;

Hilgard, P. Synthesis and Pharmacological Activity of Cyclobutane-Platinum (II) Complexes. Eur. Food Res. Technol. 1989, 22, 163. 93.

Voegeli, R.; Schumacher, W.; Engel, J.; Respondek, J.; Hilgard, P. D-19466, a

New Cyclobutane-Platinum Complex with Antitumor Activity. J. Cancer Res. Clin. Oncol. 1990, 116, 439-442. 94.

Adis R&D Profile. Lobaplatin: D 19466. Drugs R&D 2003, 4, 369-372.

ACS Paragon Plus Environment

55

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

95.

Page 56 of 66

Chen, M. Q.; Chen, C.; Lu, H. J.; Xu, B. H. The Efficacy and Toxicities of

Combined Lobaplatin with Paclitaxel as a First-Line Chemotherapy for Advanced Esophageal Squamous Cell Carcinoma. J. Thorac. Dis. 2015, 7, 1749-1755. 96.

Tanaka, K.; Kunimatsu, T.; Shimakura, J.; Hanada, M. Development of

Miriplatin, a Novel Antitumor Platinum for Hepatocellular Carcinoma; Sumitomo Kagaku R&D Reports; 2011, I1-12. 97.

Horikawa, M.; Miyayama, S.; Irie, T.; Kaji, T.; Arai, Y. Development of

Conventional Transarterial Chemoembolization for Hepatocellular Carcinomas in Japan: Historical, Strategic, and Technical Review. Am. J. Roentgenol. 2015, 205, 764-773. 98.

Nishikawa, H.; Kita, R.; Kimura, T.; Osaki, Y. Transcatheter Arterial Embolic

Therapies for Hepatocellular Carcinoma: A Literature Review. Anticancer Res. 2014, 34, 6877-6886. 99.

Maeda, M.; Uchida, N. A.; Sasaki, T. Liposoluble Platinum (II) Complexes with

Antitumor Activity. Jpn. J. Cancer Res. 1986, 77, 523-525. 100.

Monneret, C. Platinum Anticancer Drugs. From Serendipity to Rational Design.

Ann. Pharm. Fr. 2011, 69, 286-295. 101.

Lee, J. W.; Park, J. K.; Lee, S. H.; Kim, S. Y.; Cho, Y. B.; Kuh, H. J. Anti-Tumor

Activity of Heptaplatin in Combination with 5-Fluorouracil or Paclitaxel against Human Head and Neck Cancer Cells in Vitro. Anticancer Drugs 2006, 17, 377-384. 102.

Garattini, S. Are Me-Too Drugs Justified? J. Nephrol. 1997, 10, 283-294.

103.

Kennedy, I. C.; Fitzharris, B. M.; Colls, B. M.; Atkinson, C. H. Carboplatin Is

Ototoxic. Cancer Chemother. Pharmacol. 1990, 26, 232-234.

ACS Paragon Plus Environment

56

Page 57 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

104.

Chitapanarux, I.; Lorvidhaya, V.; Kamnerdsupaphon, P.; Sumitsawan, Y.;

Tharavichitkul, E.; Sukthomya, V.; Ford, J. Chemoradiation Comparing Cisplatin Versus Carboplatin in Locally Advanced Nasopharyngeal Cancer: Randomised, Non-Inferiority, Open Trial. Eur. J. Cancer 2007, 43, 1399-1406. 105.

Minami, S. B.; Sha, S. H.; Schacht, J. Antioxidant Protection in a New Animal

Model of Cisplatin-Induced Ototoxicity. Hear. Res. 2004, 198, 137-143. 106.

Jacobs, S.; Mccully, C. L.; Murphy, R. F.; Bacher, J.; Balis, F. M.; Fox, E.

Extracellular Fluid Concentrations of Cisplatin, Carboplatin, and Oxaliplatin in Brain, Muscle, and Blood Measured Using Microdialysis in Nonhuman Primates. Cancer Chemother. Pharmacol. 2010, 65, 817-824. 107.

Phosplatin therapeutics. http://phosplatin.com (July 30, 2016).

108.

Phosplatin therapeutics. A Phase 1, Open-Label, Dose-Escalation Study

Evaluating the Safety, Pharmacokinetics, and Clinical Effects of Intravenously Administered Pt-112 Injection in Subjects with Advanced Solid Tumors. https://clinicaltrials.gov/ct2/show/NCT02266745 (August 01, 2016) 109.

Mishur, R. J.; Zheng, C.; Gilbert, T. M.; Bose, R. N. Synthesis, X-Ray

Crystallographic, and NMR Characterizations of Platinum(II) and Platinum(IV) Pyrophosphato Complexes. Inorg. Chem. 2008, 47, 7972-7982. 110.

Bose, R. N.; Maurmann, L.; Mishur, R. J.; Yasui, L.; Gupta, S.; Grayburn, W. S.;

Hofstetter, H.; Salley, T. Non-DNA-Binding Platinum Anticancer Agents: Cytotoxic Activities of Platinum–Phosphato Complexes Towards Human Ovarian Cancer Cells. Proc. Natl. Acad. Sci. U. S. A. 2008, 105, 18314-18319.

ACS Paragon Plus Environment

57

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

111.

Page 58 of 66

Bose, R. N.; Moghaddas, S.; Belkacemi, L.; Tripathi, S.; Adams, N. R.;

Majmudar, P.; Mccall, K.; Dezvareh, H.; Nislow, C. Absence of Activation of DNA Repair Genes and Excellent Efficacy of Phosphaplatins against Human Ovarian Cancers: Implications to Treat Resistant Cancers. J. Med. Chem. 2015, 58, 8387-8401. 112.

Moghaddas, S.; Majmudar, P.; Marin, R.; Dezvareh, H.; Qi, C.; Soans, E.; Bose,

R. N. Phosphaplatins, Next Generation Platinum Antitumor Agents: A Paradigm Shift in Designing and Defining Molecular Targets. Inorg. Chim. Acta 2012, 393, 173-181. 113.

Businesswire. Phosplatin Therapeutics Announces the Execution of Regional

Licensing Agreement for Its Novel Therapeutic Agent in Oncology, Pt-112. http://www.businesswire.com/news/home/20150818005165/en/Phosplatin-TherapeuticsAnnounces-Execution-Regional-Licensing-Agreement (July 30, 2016) 114.

Placon Therapeutics. Placon Therapeutics Announces Company Launch and Fda

Acceptance of IND for Novel Platinum Candidate BTP-114. http://www.placontx.com/wp-content/uploads/2016/03/FDA-Acceptance-of-IND-forNovel-Platinum-Candidate-BTP-114_032316.pdf (July 29, 2016) 115.

Placon Therapeutics. http://www.placontx.com/ (July 30, 2016).

116.

Thepharmaletter. Placon Therapeutics Spun out as Independent Company.

http://www.thepharmaletter.com/article/placon-therapeutics-spun-out-as-independentcompany (July 29, 2016) 117.

Moreau, B.; Alargova, R.; Brockman, A.; Whalen, K.; Quinn, J.; Meetze, K.;

Bazinet, P.; Dupont, M.; Krawiec, B.; Kriksciukaite, K.; Lemelin, C.; Limsoo, P.; Oller, H.; Ramstack, M.; Rockwood, D.; Shinde, R.; Singh, S.; White, B.; Auyeung, T.; Dunbar, C.; Bilodeau, M.; Wooster, R. Abstract 4484: BTP-114: An Albumin Binding

ACS Paragon Plus Environment

58

Page 59 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Cisplatin Prodrug with Improved and Sustained Tumor Growth Inhibition. Cancer Res. 2015, 75, 4484-4484. 118.

Smith, M. E. B.; Caspersen, M. B.; Robinson, E.; Morais, M.; Maruani, A.;

Nunes, J. P. M.; Nicholls, K.; Saxton, M. J.; Caddick, S.; Baker, J. R.; Chudasama, V. A Platform for Efficient, Thiol-Stable Conjugation to Albumin's Native Single Accessible Cysteine. Org. Biomol. Chem. 2015, 13, 7946-7949. 119.

Pichler, V.; Mayr, J.; Heffeter, P.; Dömötör, O.; Enyedy, É. A.; Hermann, G.;

Groza, D.; Köllensperger, G.; Galanksi, M.; Berger, W.; Keppler, B. K.; Kowol, C. R. Maleimide-Functionalised Platinum(IV) Complexes as a Synthetic Platform for Targeted Drug Delivery. Chem. Commun. (Cambridge, U. K.) 2013, 49, 2249-2251. 120.

Zheng, Y. R.; Suntharalingam, K.; Johnstone, T. C.; Yoo, H.; Lin, W.; Brooks, J.

G.; Lippard, S. J. Pt(IV) Prodrugs Designed to Bind Non-Covalently to Human Serum Albumin for Drug Delivery. J. Am. Chem. Soc. 2014, 136, 8790-8798. 121.

Kundranda, M. N.; Niu, J. Albumin-Bound Paclitaxel in Solid Tumors: Clinical

Development and Future Directions. Drug Des., Dev. Ther. 2015, 9, 3767-3777. 122.

Manaenko, A.; Chen, H.; Kammer, J.; Zhang, J. H.; Tang, J. Comparison Evans

Blue Injection Routes: Intravenous Versus Intraperitoneal, for Measurement of BloodBrain Barrier in a Mice Hemorrhage Model. J. Neurosci. Methods 2011, 195, 206-210. 123.

Mcdonald, D. M.; Baluk, P. Significance of Blood Vessel Leakiness in Cancer.

Cancer Res. 2002, 62, 5381-5385. 124.

Yu, J. J.; Yang, X.; Song, Q.; Mueller, M. D.; Remick, S. C. Dicycloplatin, a

Novel Platinum Analog in Chemotherapy: Synthesis of Chinese Pre-Clinical and Clinical Profile and Emerging Mechanistic Studies. Anticancer Res. 2014, 34, 455-463.

ACS Paragon Plus Environment

59

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

125.

Page 60 of 66

Li, G.-Q.; Chen, X.-G.; Wu, X.-P.; Xie, J.-D.; Liang, Y.-J.; Zhao, X.-Q.; Chen,

W.-Q.; Fu, L.-W. Effect of Dicycloplatin, a Novel Platinum Chemotherapeutical Drug, on Inhibiting Cell Growth and Inducing Cell Apoptosis. PLoS ONE 2012, 7, e48994. 126.

Liu, K. J.; Guan, Z. Z.; Liang, Y.; Yang, X. Q.; Peng, J.; Huang, H.; Shao, Q. X.;

Wang, M. Z.; Zhu, Y. Z.; Wu, C. P.; Wang, S. B.; Xiong, J. P.; Bai, Y. X.; Yu, S. Y.; Zhang, Y.; Hu, X. H.; Feng, J. F.; Wu, S. X.; Jiao, S. C.; Zhou, C. C.; Wang, J.; Wu, H. Y. A Double-Blind, Randomized Phase II Study of Dicycloplatin Plus Paclitaxel Versus Carboplatin Plus Paclitaxel as First-Line Therapy for Patients with Advanced Non-SmallCell Lung Cancers. Arch. Med. Sci. 2014, 10, 717-724. 127.

Kang, X.; Xiao, H.-H.; Song, H.-Q.; Jing, X.-B.; Yan, L.-S.; Qi, R.-G. Advances

in Drug Delivery System for Platinum Agents Based Combination Therapy. Cancer Biol. Med. 2015, 12, 362-374. 128.

Xue, X.; Hall, M. D.; Zhang, Q.; Wang, P. C.; Gottesman, M. M.; Liang, X. J.

Nanoscale Drug Delivery Platforms Overcome Platinum-Based Resistance in Cancer Cells Due to Abnormal Membrane Protein Trafficking. ACS Nano 2013, 7, 10452-10464. 129.

Duggan, S. T.; Keating, G. M. Pegylated Liposomal Doxorubicin. Drugs 2011,

71, 2531-2558. 130.

Liu, D., He, C., Wang, A. Z., Lin, W. Application of Liposomal Technologies for

Delivery of Platinum Analogs in Oncology. Int. J. Nanomed. 2013, 8, 3309-3319. 131.

European Medicines Agency. Public Summary of Positive Opinion for Orphan

Designation of Cisplatin (Liposomal) for the Treatment of Pancreatic Cancer. Preauthorization Evaluation of Medicines for Human Use. Committee for Orphan Medicinal Products: London, November 30, 2007.

ACS Paragon Plus Environment

60

Page 61 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

132.

Han, X.; Sun, J.; Wang, Y.; He, Z. Recent Advances in Platinum (Iv) Complex-

Based Delivery Systems to Improve Platinum (II) Anticancer Therapy. Med. Res. Rev. 2015, 35, 1268-1299. 133.

Martin, R. B. Platinum Complexes: Hydrolysis and Binding to N(7) and N(1) of

Purines; In Cisplatin: Chemistry and Biochemistry of a Leading Anticancer Drug; Lippart, B., Ed.; Verlag Helvetica Chimica Acta: Zürich, Switzerland, 2006, 181-205. 134.

Kataoka, K.; Harada, A.; Nagasaki, Y. Block Copolymer Micelles for Drug

Delivery: Design, Characterization and Biological Significance. Adv. Drug Deliv. Rev. 2001, 47, 113-131. 135.

M.D. Anderson Cancer Center, NanoCarrier Co., Ltd. A Phase 1 Dose-Escalation

and Pharmacokinetic Study of Nc-4016 in Patients with Advanced Solid Tumors or Lymphoma. https://clinicaltrials.gov/ct2/show/NCT01999491 (August 01, 2016) 136.

Nanocarrier. Research and Development Pipeline.

http://www.nanocarrier.co.jp/en/research/pipeline/index.html (July 30, 2016). 137.

Nowotnik, D. P.; Cvitkovic, E. Prolindac™ (Ap5346): A Review of the

Development of an HPMA DACH Platinum Polymer Therapeutic. Adv. Drug Delivery Rev. 2009, 61, 1214-1219. 138.

Duncan, R.; Vicent, M. J. Do Hpma Copolymer Conjugates Have a Future as

Clinically Useful Nanomedicines? A Critical Overview of Current Status and Future Opportunities. Adv. Drug Delivery Rev. 2010, 62, 272-282. 139.

Carter, P. J.; Senter, P. D. Antibody-Drug Conjugates for Cancer Therapy. Cancer

J. 2008, 14, 154-169.

ACS Paragon Plus Environment

61

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

140.

Page 62 of 66

Lambert, J. M.; Chari, R. V. Ado-Trastuzumab Emtansine (T-DM1): An

Antibody-Drug Conjugate (ADC) for Her2-Positive Breast Cancer. J. Med. Chem. 2014, 57, 6949-6964. 141.

Mehmood, R. K.; Parker, J.; Ahmed, S.; Qasem, E.; Mohammed, A. A.; Zeeshan,

M.; Jehangir, E. Review of Cisplatin and Oxaliplatin in Current Immunogenic and Monoclonal Antibody Treatments. Oncol. Rev. 2014, 8, 97-108. 142.

Huang, R.; Sun, Y.; Zhang, X. Y.; Sun, B. W.; Wang, Q. C.; Zhu, J. Biological

Evaluation of a Novel Herceptin-Platinum (II) Conjugate for Efficient and Cancer Cell Specific Delivery. Biomed. Pharmacother. 2015, 73, 116-122. 143.

Lasorsa, A.; Losacco, M.; Iacobazzi, R. M.; Porcelli, L.; Azzariti, A.; Natile, G.;

Arnesano, F. Probing the Interaction between Cisplatin and the Therapeutic Monoclonal Antibody Trastuzumab. RSC Adv. 2016, 6, 29229-29236. 144.

Mei, H. E.; Leipold, M. D.; Maecker, H. T. Platinum-Conjugated Antibodies for

Application in Mass Cytometry. Cytometry, Part A 2016, 89, 292-300. 145.

Waalboer, D. C.; Muns, J. A.; Sijbrandi, N. J.; Schasfoort, R. B.; Haselberg, R.;

Somsen, G. W.; Houthoff, H. J.; Van Dongen, G. A. Platinum(II) as Bifunctional Linker in Antibody-Drug Conjugate Formation: Coupling of a 4-Nitrobenzo-2-Oxa-1,3-Diazole Fluorophore to Trastuzumab as a Model. ChemMedChem 2015, 10, 797-803. 146.

Johnstone, T. C.; Suntharalingam, K.; Lippard, S. J. The Next Generation of

Platinum Drugs: Targeted Pt(II) Agents, Nanoparticle Delivery, and Pt(IV) Prodrugs. Chem. Rev. (Washington, DC, U. S.) 2016, 116, 3436-3486.

ACS Paragon Plus Environment

62

Page 63 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

147.

Mukhopadhyay, S.; Barnes, C. M.; Haskel, A.; Short, S. M.; Barnes, K. R.;

Lippard, S. J. Conjugated Platinum(IV)-Peptide Complexes for Targeting Angiogenic Tumor Vasculature. Bioconjugate Chem. 2008, 19, 39-49. 148.

Massaguer, A.; Gonzalez-Canto, A.; Escribano, E.; Barrabes, S.; Artigas, G.;

Moreno, V.; Marchan, V. Integrin-Targeted Delivery into Cancer Cells of a Pt(IV) ProDrug through Conjugation to Rgd-Containing Peptides. Dalton Trans. 2015, 44, 202-212. 149.

Gibson, D. The Mechanism of Action of Platinum Anticancer Agents--What Do

We Really Know About It? Dalton Trans. 2009, 48, 10681-10689. 150.

Klein, A. V.; Hambley, T. W. Platinum Drug Distribution in Cancer Cells and

Tumors. Chem. Rev. 2009, 109, 4911-4920. 151.

Molenaar, C.; Teuben, J. M.; Heetebrij, R. J.; Tanke, H. J.; Reedijk, J. New

Insights in the Cellular Processing of Platinum Antitumor Compounds, Using Fluorophore-Labeled Platinum Complexes and Digital Fluorescence Microscopy. J. Biol. Inorg. Chem. 2000, 5, 655-665. 152.

Benedetti, B. T.; Peterson, E. J.; Kabolizadeh, P.; Martínez, A.; Kipping, R.;

Farrell, N. P. Effects of Noncovalent Platinum Drug–Protein Interactions on Drug Efficacy: Use of Fluorescent Conjugates as Probes for Drug Metabolism. Mol. Pharmaceutics 2011, 8, 940-948. 153.

Miller, M. A.; Askevold, B.; Yang, K. S.; Kohler, R. H.; Weissleder, R. Platinum

Compounds for High-Resolution in Vivo Cancer Imaging. ChemMedChem 2014, 9, 1131-1135. 154.

Jagodinsky, J. C.; Sulima, A.; Cao, Y.; Poprawski, J. E.; Blackman, B. N.; Lloyd,

J. R.; Swenson, R. E.; Gottesman, M. M.; Hall, M. D. Evaluation of Fluorophore-

ACS Paragon Plus Environment

63

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 64 of 66

Tethered Platinum Complexes to Monitor the Fate of Cisplatin Analogs. J. Biol. Inorg. Chem. 2015, 20, 1081-1095. 155.

Shen, C.; Harris, B. D. W.; Dawson, L. J.; Charles, K. A.; Hambley, T. W.; New,

E. J. Fluorescent Sensing of Monofunctional Platinum Species. Chem. Commun. (Cambridge, U. K.) 2015, 51, 6312-6314. 156.

Ding, S.; Qiao, X.; Suryadi, J.; Marrs, G. S.; Kucera, G. L.; Bierbach, U. Using

Fluorescent Post-Labeling to Probe the Subcellular Localization of DNA-Targeted Platinum. Angew. Chem. Int. Ed. 2013, 52, 3350-3354. 157.

White, J. D.; Haley, M. M.; Derose, V. J. Multifunctional Pt(II) Reagents:

Covalent Modifications of Pt Complexes Enable Diverse Structural Variation and in-Cell Detection. Acc. Chem. Res. 2016, 49, 56-66. 158.

White, J. D.; Osborn, M. F.; Moghaddam, A. D.; Guzman, L. E.; Haley, M. M.;

Derose, V. J. Picazoplatin, an Azide-Containing Platinum(II) Derivative for Target Analysis by Click Chemistry. J. Am. Chem. Soc. 2013, 135, 11680-11683. 159.

Osborn, M. F.; White, J. D.; Haley, M. M.; Derose, V. J. Platinum-Rna

Modifications Following Drug Treatment in S. Cerevisiae Identified by Click Chemistry and Enzymatic Mapping. ACS Chem. Biol. 2014, 9, 2404-2411. 160.

Moghaddam, A. D.; White, J. D.; Cunningham, R. M.; Loes, A. N.; Haley, M. M.;

Derose, V. J. Convenient Detection of Metal-DNA, Metal-RNA, and Metal-Protein Adducts with a Click-Modified Pt(II) Complex. Dalton Trans. 2015, 44, 3536-3539. 161.

Wirth, R.; White, J. D.; Moghaddam, A. D.; Ginzburg, A. L.; Zakharov, L. N.;

Haley, M. M.; Derose, V. J. Azide Vs Alkyne Functionalization in Pt(II) Complexes for

ACS Paragon Plus Environment

64

Page 65 of 66

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Post-Treatment Click Modification: Solid-State Structure, Fluorescent Labeling, and Cellular Fate. J. Am. Chem. Soc. 2015, 137, 15169-15175.

ACS Paragon Plus Environment

65

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 66 of 66

Table of Contents graphic

ACS Paragon Plus Environment

66