CA2720396A1 - Pharmaceutical composition - Google Patents

Pharmaceutical composition Download PDF

Info

Publication number
CA2720396A1
CA2720396A1 CA2720396A CA2720396A CA2720396A1 CA 2720396 A1 CA2720396 A1 CA 2720396A1 CA 2720396 A CA2720396 A CA 2720396A CA 2720396 A CA2720396 A CA 2720396A CA 2720396 A1 CA2720396 A1 CA 2720396A1
Authority
CA
Canada
Prior art keywords
modulator
complex
enzyme
pdh
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2720396A
Other languages
French (fr)
Inventor
Robert Shorr
Paul Bingham
Lakmal W. Boteju
Zuzana Zachar
Robert Rodriguez
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2720396A1 publication Critical patent/CA2720396A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/385Heterocyclic compounds having sulfur as a ring hetero atom having two or more sulfur atoms in the same ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

A pharmaceutically-acceptable modulator of the regulation or perturbation of the structure, expres-sion, and/or activity of at least one enzyme and/or enzyme complex, or subunit thereof, such as via the altered mito-chondrial energy metabolism of the pyruvate dehydroge-nase (PDH) complex of warm-blooded animals, including humans, and methods of use thereof, comprises an effec-tive amount of at least one lipoic acid derivative and at least one pharmaceutically-acceptable carrier thereof to af-fect the complex's phosphorylation state. By increasing PDH kinase activity and/or decreasing PDH phosphatase activity, the modulator prevents the detoxification anaero-bic glycolytic toxic metabolites through inhibition of the activity of the PDH complex's El a subunit, obliging in-creased mitochondrial oxidative phosphorylation activity.
As cells characterized by hyperproliferation, such as tumor cells, cannot also generate acetyl-CoA and NADH because of the modulator's additional action in inhibiting the action of the PDH complex's E2 subunit, the mitochondrial mem-brane polarization is lost, facilitating cell death.

Description

Pharmaceutical Composition Field of the Invention This invention relates to therapeutic and diagnostic compositions, and more particularly to pharmaceutical compositions, and methods of use thereof, which demonstrate selective uptake into cells characterized by hyperproliferation, including cancer cells, and which modulate the regulation or perturbation of the structure, expression, and/or activity of enzymes, thereby facilitating the detection and treatment or destruction of these cells. More specifically, these agents target and perturb the activity or regulation thereof of the altered mitochondrial energy metabolism observed in such locales as the modified pyruvate dehydrogenase (PDH) complex associated with most cancers.

Background of the Invention Mitochondria are the primary control centers for energy production and cellular life-and-death processes in eukaryotes. While various mechanisms of communication with the rest of the cell exist, reversible phosphorylation is an important means of regulating mitochondrial functions. (Pagliarini D.J. and Dixon J.E. (2006). Mitochondrial modulation:
reversible phosphorylation takes center stage? TRENDS in Biochem. Sci. 31:26-34, passim, herein incorporated by reference) The steadily increasing number of reported mitochondrial kinases, phosphatases, and phosphoproteins suggests that phosphorylation is likely to emerge as a common theme in the regulation of mitochondrial processes. Pathological or genetic changes associated in mitochondrial enzyme structure, function and activity regulation contribute to, and may be important targets for, the treatment of disease.

In line with their role as both a point of convergence and regulator of diverse cellular functions, mitochondria have crucial roles in apoptosis, production of reactive oxygen species (ROS), and numerous metabolic processes, including the production of more than 90% of cellular ATP. Furthermore, as cells grow and divide, new mitochondria have to be made, this process itself requiring careful coordination of nuclear and mitochondrial DNA
transcription and translation. Finally, as cellular energy needs change, mitochondria must respond rapidly by tuning their ATP output. Hence, mitochondria require a complex system of communication with cellular functions. As evident from FIGURE 1, signal molecules to and from mitochondria include ions, gases, metabolites, hormones, transcription factors and proteins. Consequently, the recognition of mitochondria as centers for receiving, integrating, and transmitting cellular signals is an important advance in the design and testing of pharmaceuticals.

A cornerstone of signalling in mitochondrial regulation is reversible phosphorylation.
However, while the first demonstration of a protein kinase event was reported in 1954, and the fact that a core mitochondrial function can be regulated by reversible phosphorylation was discovered nearly four decades ago, reports of mitochondrial phosphorylation events are scarce. This is so despite the recognition in the 1980s and 1990s of numerous signal transduction phosphorylation cascades that traverse the plasma membrane and extend through the cytosol to the nucleus. Such paucity of knowledge may be due partly to the fact that the bulk of the mitochondrial protein machinery lies behind two lipid bilayers, which seemingly places mitochondrial proteins out of the reach of cytosolic signalling cascades. In any event, it has been not widely accepted that the mitochondrion regulates signalling by reversible phosphorylation in a manner key to disease management. Nevertheless, as seen in Table 1, by 2006 more than 60 proteins in all mitochondrial compartments (i.e., the matrix, inner membrane, intermembrane space, and outer membrane, including the cytoplasmic-facing outer surface) had been identified as phosphoproteins implicated in a wide spectrum of mitochondrial functions. Mounting data further demonstrates the importance of reversible phosphorylation of mitochondrial targets and the use of compositions targeting the same for the improved treatment of cancer.

Table 1. Mitochondrial phosphoproteinse No. Protein Location P site Source Function Refs 1 PDC Eta M Ser Various Acetyl-CoA formation [11) 2 PDC E10 M Tyr Human sperm Acetyl-CoA formation (8]
3 PDC E3 M Tyr Hamster sperm Regulation of the PDC (8]
4 PDK isoform 2a M Ser/Thr Rat TCA. 1271 Aconitase M 7 Bovine/potato TCA [22) 6 NAD-isocitrate dehydrogenase M ? Bovine/potato TCA [221.
7 NAD-malete dehydrogenase M ? Potato TCA. (221 8 NAD-malic enzyme M ? Potato TCA [221 9 Succinyl-CoA-ligase x subunit M ? Rat/potato TCA [22,471 Succinyl-CoA-ligase (i subunit M 7 Rat/potato TCA [22,47) 11 Formate dehydrogenase M Ser/Thr Potato TCA [22,231 12 Aconitase M Tyr. Guinea pig synaptosomes TCA (81 13 BCKAD M Ser Various AA metabolism [311 14 BCKAD kinase M Ser Rat AA Metabolism 154]
HSP22 M Ser Corn Chaperone [391 16 HSP 90 M 7 Potato Chaperone. [22]
17 Chaperonin 60 M 7 Potato Chaperone (221 18 mthsp75 M Tyr Rat hepatome cells Chaperone [29]
19 TRAP-1` M Tyr Human sperm Chaperone [81 CYP2E1' M/IM? Ser COS Detoxification [43]
21 CYP2B10 M/IM? Ser COS Detoxification 1181 22 GSTA 4-4 M . Ser/Thr COS Detoxification (44]
23 DBP M 7 Yeast mRNAturnover [381 24 MnSOD M 7 Potato Oxidative stress defense [22) EF-Tu M Thr? Rabbit heart Protein synthesis (32]
26 Creatine kinase M 7 Bovine Synthesis of phosphocreatine [451 27 MTERFh M ? Rat Transcription termination [42]
28 Abf2p M Ser/Thr7 Yeast mtDNA maintenance [26j 29 MtTBP5 M ? Yeast mtDNA maintenance (501 NOK M/IMS Ser/His Pisumsativum Nucleoside triphosphate balance [49]
31 StAR IMS Ser COS-1 Steroid hormone synthesis [19]
32 Axmito` M? Tyr Rat Regulation of PLAs? (531 33 SSATe M? Ser? Rat Acetylation of spermidine [21]
34 Sab OM Ser Rat cardiac myocytes SH3 domain-binding protein [28]
CPT-I OM Ser Rat (t-OX [351 36 MtGAT` OM Ser/Thr? Rat Glycerolipid biosynthesis [411 37 BAD` OM Ser FLS.12 cells Apoptosis [30) 38 BCL-2 OM Ser Jurkat Apoptosis [17,521 39 BCI-XL OM Thr U-937 cells Apoptosis 136) CREB M/IM Ser? Rat mtDNA transcription? (24]
41 VDAC OM Tyr Guinea pig synaptosomes transport 18,451 42 Cl: ESSS iM Ser . Bovine OXPHOS 1251 43 Cl: 10 kDa IM Ser Bovine OXPHOS [25]
44 CI 42 kOa (2 sites) IM Ser & Thr Bovine OXPHOS [45,55) CIII core I . IM Tyr Human sperm OXPHOS (81 46 CIII core II IM ? Bovine OXPHOS- [451 47 CIV I IM. Tyr Bovine OXPHOS [371 48 CIV Il IM Tyr Osteoclasts OXPHOS [40) 49' CIV 111 IM Ser/Thr? Bovine OXPHOS [34]
CIV IV IM ? Rat OXPHOS 1461 51 CIV Vba . IM Ser/Thr? Bovine OXPHOS 134) 52 CV z IM ? Bovine/potato OXPHOS 1221 53 CV p IM Thr Human sk mus/bovine OXPHOS [331 54 CV d IM ? Potato OXPHOS [481 CV b IM 7 Potato OXPHOS 1481 56 ScIRP IM ? Rat OXPHOS 1201 57 SDH-Fp IM 7 Bovine/potato. OXPHOS 1221 58 bcl complex, [i-MPP subunit IM 7 Potato OXPHOS [22) 59 CIII core I IM Tyr Human sperm OXPHOS [8]
NAD(P) transhydrogenaso IM 7 Bovine H* pump 1451 61 ANT IM 7 Bovine Transport (451 62 Phosphate carrier protein IM 7 Bovine Transport [451 63 Aldose reductase' 7 SOFT? Various cell lines Osmoregulation [511 'Abbreviations: AA, amino acid IS, serine;T, threonine; Y, tyrosine); ANT, adenine nucleotide transporter; Axmito, mitochondria) annexin; fl-Ox, p-oxidation; BCKAD, branched chain ketoacid dehydrogenase; CI-CV. respiratory chain complexes 1-5:
CPT, carnitine palmitoyltransferese; CREB, cAMP-responsive element (Cre)-binding protein: CYP, cytochrome P450; DBP, dodecamer-binding protein; EF. elongation factor; GST. glutathione S-transterase; HSP, heat shock protein; IM. Inner membrane: IMS.
intermembrane space; M. matrix: MnSOD. manganese super oxide dismutase; mTERF, mitochondria) transcription termination factor; m1GAT. mitochondrial glycerol-phosphatase aceryltransferase; mthsp, mitochondrialHSP; mtTBP; mitochondrial telomere-binding protein; NDK, nucleoside diphosphate kinase: OM. outer' membrane:
OXPHOS; oxidative phosphorylation; P site; site of phosphoryiation: POC E1/3, pyruvate dehydrogenase complex,E1/3 subunit: PDK, pyruvate dehydrogenase kinase: PLA, phospholipaseA; ScIRP, subunit c-immunoreactive peptide; SSAT, spermidine/spermina scetyltransfarase: StAR, steroidogenic acute regulatory protein: TCA. tricarboxylie acid cycle: TRAP-1, tumor-necrosis factor type 1 receptor-associated protein:
VOAC. voltage-dependent anion channel.
Phosphorylation is observed only in.vitru on recombinant protein.
`Protein translocetes to mitochondria (i.e. protein is not a resident mitochondrial protein).
The largest kinase and phosphatase families in the human genome, the protein kinases (PKs) and protein tyrosine phosphatases (PTPs), possess more than 500 and more than 100 members, respectively. Together with smaller families of kinases and phosphatases, these signalling molecules comprise nearly three percent of all proteins encoded in the human genome. Similar to the aforementioned phosphoproteins, kinases and phosphatases have been implicated in mitochondrial functions in a surprising number of studies, and so far at least 25 kinases and eight phosphatases have been reported to localize to mitochondria, as seen in FIGURE 2. These kinases and phosphatases are clearly not restricted to one group or family; rather, they represent nearly every known mammalian kinase and phosphatase subgroup, reflecting the range of signalling pathways that are likely to influence the mitochondrion. These signalling molecules include kinases and phosphatases varying in substrate specificity, (e.g., tyrosine kinases, classic PTP subgroups, serine/threonine kinases, and dual-specific PTPs); in catalytic mechanisms (e.g., cysteine-based PTPs, aspartic acid-based PTPs, and metal-dependent phosphatases); and in evolutionary conservation (e.g., bacterially-related pyruvate dehydrogenase kinases (PDKs) and phosphatases (PDPs), branched chain ketoacid dehydrogenase kinase (BCKDK) and phosphatase (BCKDP), and many mammalian-specific enzymes).

Most of these signalling molecules possess other non-mitochondrial roles in the cell and are primarily found to exist outside mitochondria. The impetus for, or mechanism of, their translocation to mitochondria is poorly understood for most proteins.
What is clear, however, is that kinases and phosphatases, like the phosphoproteins listed earlier, are present in all compartments of the mitochondrion, as evident in FIGURE 3, and that their activities impinge on diverse mitochondrial functions.

A few signalling molecules, however, seem to localize primarily to mitochondria. In addition to PDKs and PDPs, this group includes the PTEN-induced kinase PINK 1, the dual-specific PTP targeted to the mitochondrion PTPMT 1, and the aspartic-acid-based phosphatase/ATPase Tim50. Although the substrates for many of these proteins are currently unknown, it is clear from biological and genetic data that they possess crucial functions in mitochondria. For example, although its submitochondrial localization remains to be 5 determined, P1NK1 is targeted to mitochondria by an N-terminal signal sequence. This kinase, which shares high sequence homology with the Ca+2/calmodulin-regulated kinase family, seems to be involved in pro-survival activities. Similarly, PTPMTI was recently identified as the first PTP that is localized primarily inside the mitochondrion and, like PINK1, is targeted to the mitochondrion by an N-terminal signal peptide and is found tightly associated with the matrix face of the inner mitochondrial membrane. PTPMT 1 is highly expressed in pancreatic B cells, whose mitochondria have the important function of coupling glucose metabolism to the secretion of insulin. Finally, Tim50, a key component of the TIM
(translocase of the inner membrane) complex, has sequence homology to the CTD
family of aspartic-acid-based phosphatases/ATPases. Tim50, like other members of this family, might function as an ATPase, but it has been also shown to possess phosphatase activity against the phosphotyrosine analog para-nitrophenyl phosphate in vitro. Given the above, it can be seen that not only are kinases and phosphatases recruited to mitochondria from elsewhere in the cell, but the mitochondrion itself seems to possess a contingent of resident signalling molecules.

Although the effect of phosphorylation on most known mitochondrial phosphoproteins is unclear, with the kinases and phosphatases responsible sometimes remaining unidentified, a few phosphorylation events have been partially characterized.
These examples, like the phosphoproteins and signalling molecules discussed earlier, are not restricted to one area of the mitochondrion.
Phosphorylation on the mitochondrial outer membrane has a crucial role in regulating apoptosis. A particularly well-defined event is the phosphorylation of BAD, a proapoptotic member of the BCL-2 family. It has been shown that PKA, after treatment with the pro-survival cytokine interleukin-3, translocates to the outer membrane. Once anchored to an A-kinase anchoring protein (AKAP) on the outer membrane, PKA phosphorylates BAD
on Ser 112, contributing to the inactivation and disassociation of BAD from mitochondria, a process depicted in FIGURE 3A. Phosphorylation of BAD on Ser 136 by p70S6 kinase and on Ser 155 by in unidentified kinase has also been implicated in the inactivation of BAD.

The most well-established example of reversible phosphorylation acting as a regulatory mechanism in healthy cell mitochondria is that of the PDH complex in the matrix, a simplified cartoon of which is illustrated in FIGURE 3B. This complex catalyzes the conversion of glycolysis-derived pyruvate to acetyl-coenzyme A (CoA), the main precursor to the tricarboxylic acid (TCA) cycle. As the link between these two major energy-producing pathways, the PDH complex must be properly regulated for the maintenance of cellular glucose homeostasis.

Since its identification as the first mitochondrial phosphoprotein, the PDH
complex and its regulation by reversible phosphorylation have been studied exhaustively.
Phosphorylation and dephosphorylation of the PDH complex are carried out by PDKs and PDPs, respectively. At least four PDK isoforms and two PDP isoforms are known, all of which are associated with the E2 subunit of the PDH complex. The phosphorylation events occur on three separate serine residues of the El subunit, each leading to significant inactivation of this complex. Notably, there is now at least one report of a PDK itself being phosphorylated. This phosphorylation, carried out by PKC, has been shown to inactivate PDK, potentially demonstrating an additional level of PDH complex regulation by reversible phosphorylation. Thus, the PDH complex is a prime example of mitochondria using phosphorylation to add a level of regulation to an otherwise conserved process.

As indicated by the mitochondrial tyrosine kinases and phosphatases listed earlier, phosphorylation within this organelle is not limited to serine and threonine residues. An example of tyrosine phosphorylation that affects mitochondrial energetics is seen in the regulation of cytochrome c oxidase (COX) in the inner membrane, depicted in FIGURE 3C.
COX, as the terminal enzyme in the respiratory chain, coordinately reduces oxygen to water while pumping protons across the inner membrane. Similar to the PDH complex, COX is allosterically regulated by ATP and ADP, as well as the thyroid hormone T2 and possibly Ca+2 ions. In addition to these forms of regulation, it has been shown that COX becomes phosphorylated in a cAMP-dependent manner both in vitro and in HepG2 cells in vivo. COX
comprises thirteen subunits and has been crystallized as a dimer. The phosphorylation site has been identified as Tyr 304 of subunit 1, which is located at the dimer interface on the intermembrane. The phosphorylation event markedly inhibits COX activity, perhaps by disrupting dimer formation.

In a second example of tyrosine phosphorylation of COX, a portion of the non-receptor tyrosine kinase c-Src, similar to the Lyn tyrosine kinase localizes inside mitochondria and leads to tyrosine phosphorylation of COX on an unidentified site of subunit II in osteoclasts. The result of the phosphorylation event is opposite to that seen for subunit I, leading to enhanced COX activity.

An important aspect of mitochondrial signalling is how kinases and phosphatases are themselves regulated. Numerous kinases that primarily reside elsewhere in the cell but become targeted to the mitochondrion, such as Abl, Akt, GSK3I3 and PKC6, seem to do so only in their active state. Thus, the extent of some kinase activities within mitochondria might simply be dictated by the number of enzymes that are imported into the organelle.
For resident signalling molecules, however, different regulatory means must be in place. Although these processes remain to be determined, it is likely that second messengers will have a key role. The activities of the PDK and PDP isoforms are known to be controlled by ions and small molecules such as Mgt+, Cat+, K+ and ADP. The characterization of mitochondrial nitric oxide synthases and the recent discovery of a soluble adenylate cyclase in mitochondria provide further opportunities for second messengers to contribute to regulating mitochondrial signalling molecules. Finally, ROS, which have been established as a means of regulating signalling molecules elsewhere in the cell, will almost certainly be involved in regulating kinases and phosphatases in mitochondria, where the bulk of reactive oxygen species is produced. Relative expression levels of isoforms of kinase and phosphatase may play an important role in pathology and linked to other signal transduction events associated with disease. Changes at the gene and expression level may also correlate with such changes.

Even after several thorough proteomic surveys, it is estimated that only two-thirds of the mammalian mitochondrial proteome is known. Much of the remaining third is likely to be comprised of low-abundance proteins, such as signalling proteins, which were below the detection level of these mass spectrometric analyses. What is also clear from these studies is the high variability in protein content among mitochondria from different tissues. For example, it has been found that only -50% of the proteins in their proteomics effort were conserved across the four tissues examined (i.e., brain, heart, liver and skeletal muscle). It is likely that different mitochondrial signalling pathways not only will vary from tissue to tissue in the same way but might very well contribute to this observed mitochondrial diversity.

Nonetheless, there is more than sufficient evidence to conclude that reversible phosphorylation is involved in the regulation of mitochondrial processes. With over 60 reported phosphoproteins, 30 kinases and phosphatases, and various auxiliary signalling proteins, the mitochondrion is certainly an underappreciated site for signalling by reversible phosphorylation, and in fact such regulation may be useful for the treatment of hyperproliferative diseases such as cancer.

The vast majority of fast-growth tumor cells exhibits profound genetic, biochemical, and histological differences with respect to nontransformed cells. Many of these are associated with altered energy metabolism in comparison to the tissue of origin. The most notorious and well-known energy metabolism alteration in tumor cells is an increased glycolytic capacity even in the presence of a high 02 concentration, a phenomenon known as the Warburg effect.

Warburg originally proposed that the driving force of the enhanced glycolysis in tumor cells was the energy deficiency caused by an irreversible damage of the mitochondrial function in which, similarly to anaerobic muscle, glucose is converted through glycolysis to lactate, which is later secreted. It has been proposed that this increase in the glycolytic flux in tumor cells is a metabolic strategy to ensure survival and growth in environments with low 02 concentrations, such as the partial hypoxia observed in poorly-oxygenated solid tumors.
In particular, since the concentration of 02 is lower than 20 M in many human hypoxic tumors, oxidative phosphorylation is limited therein. Consequently, glycolysis seems to be the main energy pathway in solid tumors (e.g., slow-growing melanomas and mammary adenocarcinoma).

A proportional relationship between the rate of cellular proliferation and the rate of ATP supply has been established for fast-growth tumor cells. Some authors have proposed that the glycolytic activity correlates with the degree of tumor malignancy, so that the glycolytic rate is greater in highly de-differentiated and fast-growing tumors than in slower-growing tumors or normal cells. In fact, a high level of lactate has been proposed as a predictor of malignancy. That these events are linked to additional signal transduction events and genetic changes is likely and examples include hypoxic inducing factor and the production and release of angiogenic factors.

As depicted in FIGURE 4, for years, the tricarboxylic acid (TCA) cycle was regarded as only being biologically significant only for its role in the production of ATP as an energy 5 source for the organism. However, recent studies have shown that TCA cycle activity also affects signal transduction pathway functions, including cell growth and apoptosis decisions, and that the pertinent glycolytic and TCA cycle enzymes are able to be upregulated or down-regulated. There is also a direct correlation between tumor progression and the activities of the glycolytic enzymes hexokinase and phosphofructokinase (PFK) 1, which are greatly 10 increased in fast-growth tumor cells. Accordingly, it has been postulated that tumor cells which exhibit deficiencies in their oxidative capacity are more malignant than those that have an active oxidative phosphorylation. No matter whether under hypoxic or aerobic conditions, then, cancer tissue's reliance on glycolysis is associated with increased malignancy.

The role of lipoic acid in the PDH complex of healthy cells has been well studied.
The PDH complex has three central subunits, El, E2, and E3 (pyruvate dehydrogenase, dihydrolipoyl transacetylase, and dihydrolipoamide dehydrogenase, respectively). These complexes have a central E2 core, with the other subunits surrounding this core to form the complex. In the gap between these two subunits, the lipoyl domain ferries intermediates between the active sites. The lipoyl domain itself is attached by a flexible linker to the E2 core. Upon formation of a hemithioacetal by the reaction of pyruvate and thiamine pyrophosphate, this anion attacks the S I of an oxidized lipoate species that is attached to a lysine residue. Consequently, the lipoate S2 is displaced as a sulfide or sulfhydryl moiety, and subsequent collapse of the tetrahedral hemithioacetal ejects thiazole, releasing the TPP
cofactor and generating a thioacetate on the S I of the lipoate. At this point, the lipoate-thioester functionality is translocated into the E2 active site, where a transacylation reaction transfers the acetyl from the "swinging arm" of lipoate to the thiol of coenzyme A. This produces acetyl-CoA, which is released from the enzyme complex and subsequently enters the TCA cycle. The dihydrolipoate, still bound to a lysine residue of the complex, then migrates to the E3 active site, where it undergoes a flavin-mediated oxidation back to its lipoate resting state, producing FADH2 (and ultimately NADH) and regenerating the lipoate back into a competent acyl acceptor. Should this lipoate species be interrupted, then, there would be no flow of electrons to FADH2 or generation of acetyl-CoA, and, as a consequence, a toxic buildup of pyruvate within the cell. In cancer cells the production of acetoin has been suggested to be a requirement for cellular detoxification and survival.

As stated previously, the activity of the PDH complex in mitochondria is highly regulated by a variety of allosteric effectors and by covalent modification.
PDH activity is regulated by its state of phosphorylation, being most active in the dephosphorylated state.
PDH phosphorylation is catalyzed by PDK. PDK activity is enhanced by an increase in the level of ATP, NADH, and acetyl-CoA. Negative effectors of PDK are ADP, NAD+, CoA-SH, and pyruvate, the levels of which increase when ATP levels fall. While the regulation of PDP, the enzyme which activates PDH through dephosphorylation, is not completely understood, it is known that Mg+2 and Ca+2 activate PDP.

Two products of the complex, NADH and acetyl-CoA, are negative allosteric effectors of PDH-a, the dephosphorylated active form of PDH. These effectors reduce the affinity of the enzyme for pyruvate, thus limiting the flow of carbon through the PDH

complex. In addition, NADH and acetyl-CoA are powerful positive effectors of PDK, the enzyme that inactivates PDH by converting it to the phosphorylated PDH-b form.
Since NADH and acetyl-CoA accumulate when the cell energy charge is high, it is not surprising that high ATP levels also up-regulate PDK activity, reinforcing down-regulation of PDH

activity in energy-rich cells. However, since pyruvate is a potent negative effector of PDK, when pyruvate levels rise, PDH-a will be favored even with high levels of NADH
and acetyl-CoA.

Concentrations of pyruvate which maintain PDH-a are sufficiently high so that, in ATP-rich cells, the allosterically down-regulated, high Km form of PDH is nonetheless capable of converting pyruvate to acetyl-CoA. With large amounts of pyruvate in cells having high ATP and NADH levels, pyruvate carbon will be directed to the two main storage forms of carbon (glycogen via gluconeogenesis and fat production via fatty acid synthesis) where acetyl-CoA is the principal carbon donor. Although the regulation of PDP-b is not well understood, it is quite likely regulated to maximize pyruvate oxidation under diminished ATP concentrations and to minimize PDH activity under high ATP concentrations.

Tumor cells cannot indefinitely build up pyruvate and associated aldehydes and radicals, such as acetaldehyde, superoxide, hydrogen peroxide, and hydroxyl radicals, as these molecules are cytotoxic at high levels through such mechanisms as drastically lowering cellular pH. It has thus been described, for AS-30D and Ehrlich hepatomas, that a significant fraction of mitochondrial pyruvate is decarboxylated to an active acetaldehyde by the El component of the PDH complex via bound (3-hydroxyethylthiamine pyrophosphate.
This active acetaldehyde is in turn condensed with a second acetaldehyde, ultimately deacidifying or reducing the original pyruvate, by using either the amino acid glutamine or lipoic acid, to generate acetoin (3-hydroxybutanone), a compound which both competitively inhibits PDH

and which is less toxic to the cell than its pyruvate precursor (e.g., by maintaining pH
homeostasis within the cell). Despite the importance of acetoin in the pathway of tumor cell detoxification as a result of pyruvate buildup due to the tumor cell's reliance of glycolysis as a source of ATP production, however, there is little reference in the prior art to the effects of blocking the production of acetoin on tumor cell viability.
Recent studies suggest that forcing cancer cells into more aerobic metabolism suppresses tumor growth. The transition to Warburg metabolism therefore requires shutting down the PDH complex. In this transition, there is enhanced signalling by hypoxia-inducing factor (HIF) in cancer cells, not surprising given HIF's significant role in the metabolism of glucose, as shown in FIGURE 5. Mutations that directly or indirectly instigate HIF signalling in fact appear to be a common mechanism in the development of cancer. HIF
induces the overexpression of PDK1, which then acts to lower PDH complex activity.
Phosphorylation by PDK1 can be particularly effective for maintaining an inactive PDH complex since this isoform uniquely phosphorylates three serine residues in the alpha subunit of El, the first subunit of the PDH complex. Reactivation of El requires the removal of all three phosphate groups. Furthermore, PDH complex activation may lead to the enhanced ROS
production, which may in turn lead to apoptosis. However, alterations in PDK1 observed in cancer may not only be due to changes in its concentration but also to changes in its activity and possibly in its amino acid sequence, even between one tumor type or one patient to another.

Additionally, PDK1 may form different complexes with various molecules associated with tumors depending upon tumor type. Thus, inhibition of PDK may be a potential target in generating apoptosis in tumors. However, to date, known PDKI inhibitors have been demonstrated to cause maximally only 60% inhibition of this isozyme.

While traditional chemotherapy targets dividing, proliferating cells, all clinically-accepted chemotherapeutic treatments use large drug doses that also induce profound damage to normal, proliferative host cells. Therefore, more selective targeting is required for the treatment of cancer. Another problem associated with chemotherapy is that, in many tumor types, there is either inherent or acquired resistance to antineoplastic drugs. Overall, traditional chemotherapy currently offers little long-term benefit for most malignant tumors and is often associated with adverse side-effects that diminish the length or quality of life.
Hence, radical new approaches are required that can provide long-term management of tumors while permitting a decent quality of life.

Certainly, drug efficacy, delivery, and side-effects are problems that need to be solved in developing new chemotherapies. In solid tumors, delivery to a hypoxic region may be difficult when the drug does not permeate through the different cellular layers easily. To eliminate these uncertainties, it seems relevant to design anticancer agents having metabolic inhibition constants in, at least, the submicromolar range. It may be argued that cancer cells are genetic and phenotypically heterogeneous from line to line. However, all tumor cell lines depend on glycolysis and oxidative phosphorylation for ATP supply.
Concentrating on the Warburg effect allows for designing drugs based on the physico- and biochemical energetic differences between tumor and normal cells to facilitate the design of delivery and therapeutic strategies that selectively affect solely tumor metabolism and growth, without affecting healthy host tissue and organ functionality.

US Patents 6,331,559 and 6,951,887 to Bingham et al., as well as US
Provisional Application No. 60/912,598 by Bingham et al., all herein incorporated by reference, disclose a novel class of lipoic acid derivative therapeutic agents which selectively target and kill both tumor cells and certain other types of diseased cells. These patents further disclose pharmaceutical compositions, and methods of use thereof, comprising an effective amount of such lipoic acid derivatives along with a pharmaceutically acceptable carrier.
However, while these patents describe the structures of and general use for these lipoic acid derivatives, there is no indication in either patent that these derivatives are useful in modulating the structure and/or expression level, and/or regulating the activity, of the PDH
complex.

As it has been demonstrated that the structure and/or activity of the PDH
complex is a critical determinant of tumor activity, then, it would be beneficial to provide for a PCT/US08/04410 09-09-2008 PFf lU .jZsRM8ORM 93AO10 pharmaceutically-acceptable modulator of the phosphorylation state of the PDH
complex, and methods of use thereof.

Obiects of the Invention and Industrial Applicability 5 Consequently, it is an object of the present invention to provide a pharmaceutical composition to be used in the treatment or diagnosis of a disease, condition, or syndrome characterized by cellular hyperproliferation, such as cancer, which exhibits selective activity in tumor cells.

It is a further object of the present invention to provide a pharmaceutical composition 10 to be used in the treatment or diagnosis of such an aforementioned disease, condition, or syndrome which causes minimal side effects upon administration.

It is a still further object of the present invention to provide a pharmaceutical composition to be used in the treatment or diagnosis of such an aforementioned disease, condition, or syndrome which is easily manufactured at the least possible cost and is capable 15 of being stored for the longest possible period.

It is a still further object of the present invention to provide a pharmaceutical composition to be used in the treatment or diagnosis of such an aforementioned disease, condition, or syndrome which modulates mitochondrial energy metabolism, especially via the phosphorylation state of the PDH complex in tumor cell mitochondria.

Summary of the Invention To achieve the aforementioned aims, the present invention broadly provides a pharmaceutical composition useful for treating, diagnosing, or preventing a disease, condition, or syndrome characterized by an alteration of the phosphorylation state of at least one enzyme and/or enzyme complex, or subunit thereof, such as the PDH complex, including AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 FF FfTI%WMU441 t' *Q1&P' 0 those characterized by cellular hyperproliferation, such as cancer, or symptoms thereof, in warm-blooded animals, including humans, wherein the pharmaceutical composition comprises an effective amount of at least one lipoic acid derivative, including those as described in US Patents 6,331,559 and 6,951,887 and US Provisional Application No.

60/912,598, all herein incorporated by reference, and at least one pharmaceutically-acceptable carrier thereof.

By inhibiting mitochondrial energy metabolism, the lipoic acid derivatives of the present invention cause both the loss of mitochondrial membrane potential and other mitochondrial consequences in the diseased cell, resulting in the irreversible initiation of cell death. The lipoic acid derivatives of the present invention may also inhibit mitochondrial energy metabolism by the activation of PDKs and/or inhibition of PDPs or by inhibiting the conversion of pyruvate to the less-toxic molecule acetoin through inhibition of the activity of the El subunit of the PDH complex. The inhibition of acetoin synthesis will distort other processes, including redox balance and may also cause the production of toxic by-products, including acetaldehyde, superoxide, hydrogen peroxide, and hydroxyl radical, these by-products themselves consequently causing irreversible damage to the mitochondrion of the diseased cell.

The pharmaceutical composition of the present invention may modulate the effects of PDKI, PDK2, PDK3, PDK4, and the mutants or isoforms of each thereof. The pharmaceutical compound may also modulate the effects of PDP1, PDP2 and the isoforms of each thereof.

The pharmaceutical composition of the present invention may also modulate the expression level of the phosphorylase, kinase, and dehydrogenase enzyme constituents found in the PDH complex. This modulation may occur at the transcriptional, translational, or post-translational stage, including epigenetic silencing of the appropriate genes.

AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 PiTufflZUBORN %.%S- 2&0 0 O
As a compound derived from a molecule fundamentally associated with the TCA
cycle, and by extension glycolysis, the pharmaceutical composition of the present invention demonstrates selective uptake into tumor cells. Furthermore, such selective tumor cell uptake minimizes the side effects the administration of this pharmaceutical composition would have on healthy non-transformed cells and tissue.

In one embodiment of the present invention, the lipoic acid derivatives have the general formula (I):

S

(CH2)x R3 (1) wherein R, and R2 are independently selected from the group consisting of hydrogen, alkyl CnH2õ+i, alkene Cõ H2,,, alkenyl Cõ H2i_,, alkyine CõH2õ-2, alkynyl G,H2r_3, alkyl sulfide CH3(CH2)n-S-, disulfide alkyl CH3CHr-S-S-, thiocarbamic ester (CH2)õ C=NH-, and semithioacetal CH3CH(OH)--S-, wherein n is 1-10 and t is 0-9, aromatic, acyl defined as R4C(O)-, heteroaryl, imidoyl defined as R5C(=NH)-, organometallic aryl, alkyl-organometallic aryl, semiacetal R6CH(OH)-S-, amino acids, carbohydrates, nucleic acids, lipids, and multimers and combinations thereof;

wherein R, and R2 as defined above can be unsubstituted or substituted;

wherein R3 is selected from a group consisting of amino acids, carbohydrates, nucleic acids, lipids, and multimers thereof;

wherein R4 is selected from the group consisting of hydrogen, alkenyl, alkynyl, alkylaryl, heteroaryl, alkylheteroaryl and organometallic aryl, any of which can be substituted or unsubstituted;

wherein R5 is selected from the group consisting of hydrogen, alkenyl, alkynyl, aryl, alkylaryl, heteroaryl, and alkylheteroaryl, any of which can be substituted or unsubstituted;
AMENDED SHEET - IPEA/US
wherein R6 is CC13, CF3, or COON;

and wherein x is 0-16;
or salts thereof.

In a second embodiment of the present invention, the lipoic acid derivatives are defined by a second general formula (II):

CI(CH2)x R3 (II) wherein M is a covalent bond, -[C(R,)(R2)]Z , or a metal chelate or other metal complex where the metal is not palladium;

wherein R, and R2 are independently selected from the group consisting of hydrogen, acyl R4C(O)-, alkyl CõH2õ+,, alkenyl defined as C,,,H2m4, alkynyl defined as CmH2m-3, aryl, heteroaryl, alkyl sulfide CH3(CH2)õ--S-, imidoyl defined as R4C(=NH)-, hemiacetal defined as R6CH(OH)-S-, amino acids, carbohydrates, nucleic acids, lipids, and multimers and combinations thereof, wherein R, and R2 as defined above can be unsubstituted or substituted;

wherein R3 is selected from a group consisting of amino acids, carbohydrates, nucleic acids, lipids, and multimers thereof;

wherein R4 and R5 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, alkylaryl, heteroaryl, and heterocyclyl, any of which can be substituted or unsubstituted;

wherein R5 is selected from the group consisting of CCl3, CF3 or COOH;
and wherein x is 0-16, z is 0-5, n is 0-10 and m is 2-10;

or salts thereof.

AMENDED SHEET - IPEA/US

PCT/L1S08/04410 09-09-2008 PCRM gnn MUO
Ainjdj9.D9:Zd8OjO
Furthermore, as any or all of these general structures may be metabolized within the cell or mitochondrion, it is expressly intended that metabolites of the above-referenced structures are within the scope of the present invention.

In a further aspect of the present invention, there is provided a method of diagnosing, treating, or preventing a disease, condition, syndrome, or symptoms thereof, which includes an alteration of the phosphorylation state of at least one enzyme and/or enzyme complex, or subunit thereof, such as the PDH complex, including those characterized by cellular hyperproliferation, such as cancer, in warm-blooded animals, including humans, wherein the method comprises administering to such an animal an effective amount of the pharmaceutical composition disclosed herein.

In a still further aspect of the present invention, there is provided a method of diagnosing and predicting benefit in a patient presenting symptoms of a disease, condition, or syndrome, or symptoms thereof, which includes an alteration of the phosphorylation state of at least one enzyme and/or enzyme complex, or subunit thereof, such as the PDH
complex, including those characterized by cellular hyperproliferation, such as cancer, comprising obtaining a sample of cells from the patient, administering an effective amount of the pharmaceutical composition of the present invention to the cells in vitro, and obtaining the results therefrom.

Brief Description of the Figures The following drawings are illustrative of embodiments of the invention and are not intended to limit the scope of the application as encompassed by the entire specification and claims.

FIGURE 1 depicts general signal transduction molecules targeted both into and out from mitochondria and the effects thereof.

AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 AR/02n VUtS/MW 09.K&P10 FIGURE 2 shows a list of mitochondrial kinases and phosphatases and the locations within the mitochondrion thereof.

FIGURE 3A illustrates the phosphorylation of BAD by PKA in the outer mitochondrial membrane, and the effects of reversible phosphorylation thereupon.

5 FIGURE 3B presents the conversion of pyruvate into acetyl-CoA in the mitochondrial matrix by the action of the PDH complex, and the effects of reversible phosphorylation thereupon.

FIGURE 3C shows the action of COX in reducing oxygen to water and pumping protons across the inner mitochondrial membrane, and the effects of reversible 10 phosphorylation thereupon.

FIGURE 4 illustrates the structures of substrates and products in the glycolytic production of pyruvate, also showing ATP and NADH generation and associated enzymes.
FIGURE 5 shows the regulation of glucose metabolism by HIF-1.

FIGURE 6A illustrates the difference in energy metabolism between normal tissue 15 and cancer tissue in vivo.

FIGURE 6B depicts the differences between the biogenic forms of lipoic acid in the PDH complex and the lipoic acid derivatives forming part of the pharmaceutical composition of the present invention.

FIGURE 6C presents the regulation of the PDH complex by lipoyl residue effects on 20 PDK.

FIGURE 7 shows the effects of the pharmaceutical composition of the present invention on xenograft tumor growth.

FIGURE 8 shows the effect of treatment with the pharmaceutical composition of the present invention on three tumor cell types and a non-transformed cell.

AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 P TN'MUNA 0 ;9:98&010 FIGURE 9A shows ATP levels in lung cancer cells after treatment with the pharmaceutical composition of the present invention at or above the lethal threshold.

FIGURE 9B compares the pharmaceutical composition of the present invention's inhibition of ATP synthesis in pyruvate-containing media versus glucose-containing media.

FIGURE 9C compares the pharmaceutical composition of the present invention's inhibition of ATP synthesis in breast cancer cells to that in normal breast cells.

FIGURE 9D compares the pharmaceutical composition of the present invention's inhibition of ATP synthesis with that of lipoic acid and an inactive form of the present invention in lung cancer cells.

FIGURE 10 illustrates the pharmaceutical composition of the present invention's effects on tumor cell mitochondrial levels of the PDH complex and alpha-ketoglutarate (aKDH) dehydrogenase enzymatic activities.

FIGURE I 1A shows Western analyses of two-dimensional gels of extracts from lung cancer cells treated or mock-treated with the pharmaceutical composition of the present invention.

FIGURE 11 B shows enlargements of paired two-dimensional gel samples treated and mock-treated with the pharmaceutical composition of the present invention.

FIGURE 12A depicts the regulatory role of PDKs as modulated by endogenous lipoate covalently bound to the PDH complex E2 subunit.

FIGURE 12B depicts a possible mechanism for differential inactivation of tumor cell PDH complex by the pharmaceutical composition of the present invention.

FIGURE 13 presents the effects of the pharmaceutical composition of the present invention on mitochondrial membrane potential in H460 lung cancer cells.

FIGURE 14 shows Western blot analysis results wherein cell death pathways in diverse tumor cell types by the pharmaceutical composition of the present invention.
AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 F PCR'V99 N 0 Mi t IM . 9 a O
Detailed Description of the Invention The present invention is generally directed to pharmaceutical compositions for treating, diagnosing, or preventing a disease, condition, or syndrome, or symptoms thereof, which includes an alteration of the phosphorylation state of at least one enzyme and/or enzyme complex, or subunit thereof, such as the PDH complex, including those characterized by cellular hyperproliferation, such as cancer, or symptoms thereof, in warm-blooded animals. Such animals include those of the mammalian class, such as humans, horses, cattle, domestic animals including dogs and cats, and the like, subject to disease and other pathological conditions and syndromes characterized by cellular hyperproliferation, including cancer. The pharmaceutical composition of the present invention comprises an effective amount of at least one lipoic acid derivative, including those described in US
Patents 6,331,559 and 6,951,887 and US Provisional. Application No. 60/912,598, also known as a thioctan, and a pharmaceutically-acceptable carrier or excipient therefor. As a molecule which is not only a derivative of one which is found normally within mitochondria but also one which is instrumental to the increased glycolytic activity of tumor cells as seen' in the Warburg effect, the lipoic acid derivatives of the present invention are particularly well-suited for the selective delivery into and effective concentration within the mitochondria of cells and tissues characterized by hyperproliferation, such as tumorous ones, thereby sparing normal cells and tissue from the effects of the composition.

The pharmaceutical composition of the present invention may modulate the effects of PDKI, PDK2, PDK3, PDK4, and the isoforms of each thereof via reversible phosphorylation.
The pharmaceutical composition may also modulate the effects of PDP1, PDP2, and the isoforms and/or mutants of each thereof also by reversible phosphorylation.
Such modulation may occur through either promotion or inhibition of kinase or phosphatase activity.

AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 P,6 Y 'QUU7o6%g16lA.Di:Raoiol o By inhibiting mitochondrial energy metabolism, the lipoic acid derivatives of the present invention cause both the loss of mitochondrial membrane potential and other mitochondrial consequences in the diseased cell, resulting in the irreversible initiation of cell death. The lipoic acid derivatives of the present invention may also inhibit mitochondrial energy metabolism by the activation of PDKs and/or inhibition of PDPs or by inhibiting the conversion of pyruvate to the less-toxic molecule acetoin through inhibition of the activity of the El subunit of the. PDH complex. The inhibition of acetoin synthesis will distort other processes, including redox balance and may also cause the production of toxic by-products, including acetaldehyde, superoxide, hydrogen peroxide, and hydroxyl radical, these by-products themselves consequently causing irreversible damage to the mitochondrion of the diseased cell.

In a first embodiment of the present invention, the lipoic acid derivatives are defined by a first general formula (1):

I I
S
(CH2)x R3 (I) wherein R, and R2 are independently selected from the group consisting of hydrogen, alkyl CõH2,,+i, alkene Cõ H2,,, alkenyl CõHZõ_i, alkyne CõH2r-2, alkynyl Cõ
H2,,.3, alkyl sulfide CH3(CH2)õ-S-, disulfide alkyl CH3CH1---S--S-, thiocarbamic ester (CH2)õC=NH-, and semithioacetal CH3CH(OH}-S-, wherein n is 1-10 and t is 0-9, aromatic, acyl defined as R4C(O)-, heteroaryl, imidoyl defined as R5C(=NH)-, organometallic aryl, alkyl-organometallic aryl, semiacetal R6CH(OH)-S-, amino acids, carbohydrates, nucleic acids, lipids, and multimers and combinations thereof;

wherein R, and R2 as defined above can be unsubstituted or substituted;
AMENDED SHEET - IPEA/US

PCT/-JS08/04410 09-09-2008 FP T60&W%%i$' .0S.UAO10 wherein R3 is selected from a group consisting of amino acids, carbohydrates, nucleic acids, lipids, and multimers thereof;

wherein R4 is selected from the group consisting of hydrogen, alkenyl, alkynyl, alkylaryl, heteroaryl, alkylheteroaryl and organometallic aryl, any of which can be substituted or unsubstituted;

wherein R5 is selected from the group consisting of hydrogen, alkenyl, alkynyl, aryl, alkylaryl, heteroaryl, and alkylheteroaryl, any of which can be substituted or unsubstituted;
wherein R6 is CC13, CF3, or COOH;

and wherein x is 0-16;
or salts thereof.

In a second embodiment of the present invention, the lipoic acid derivatives are defined by a second general formula (1I):

SIM,S

(CH )x R
2 s (II) wherein M is a covalent bond, -[C(R,)(R2)]Z-, or a metal chelate or other metal complex where the metal is not palladium;

wherein R, and R2 are independently selected from the group consisting of hydrogen, acyl R4C(O)-, alkyl CõH2õ+1, alkenyl defined as CmH2m.,, alkynyl defined as CmH2m_3i aryl, heteroaryl, alkyl sulfide CH3(CH2)õ-S-, imidoyl defined as R4C(=NH)-, hemiacetal defined as R6CH(OH)-S-, amino acids, carbohydrates, nucleic acids, lipids, and multimers and combinations thereof;

wherein R, and R2 as defined above can be unsubstituted or substituted;

wherein R3 is selected from a group consisting of amino acids, carbohydrates, nucleic acids, lipids, and multimers thereof;

AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 PRIOR) G6QW 1 1 A:2doiO1 O

wherein R4 and R5 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, alkylaryl, heteroaryl, and heterocyclyl, any of which can be substituted or unsubstituted;

wherein R5 is selected from the group consisting of CC13r CF3 or COOH;
5 and wherein x is 0-16, z is 0-5, n is 0-10 and m is 2-10;

or salts thereof.

Furthermore, as any or all of these general structures may be metabolized within the cell or mitochondrion, it is expressly intended that metabolites of the above-referenced structures are within the scope of the present invention.

10 The pharmaceutical composition of the present invention may also modulate the expression levels of the phosphatase, kinase, and dehydrogenase enzyme constituents found in the PDH complex. This modulation may occur at the transcriptional, translational, or post-translational stage, including epigenetic silencing of the appropriate genes.

The compositions of the present invention may further include a pharmaceutically-15 acceptable carrier or excipients. Examples of pharmaceutically-acceptable carriers are well known in the art and include those conventionally used in pharmaceutical compositions, such as, but not limited to, antioxidants, buffers, chelating agents, flavorants, colorants, preservatives, absorption promoters to enhance bioavailability, antimicrobial agents, and combinations thereof. The amount of such additives depends on the properties desired, 20 which can readily be determined by one skilled in the art.

The pharmaceutical compositions of the present invention may routinely contain salts, buffering agents, preservatives, and compatible carriers, optionally in combination with other therapeutic ingredients. When used in medicine, the salts should be pharmaceutically acceptable, but non-pharmaceutically-acceptable salts may conveniently be used to prepare 25 pharmaceutically-acceptable salts thereof and are not excluded from the scope of the AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 FPcFtTU "YZ,uVUnIOR W:09:NO$O"
invention. Such pharmacologically- and pharmaceutically-acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, palicylic, p-toluene sulfonic, tartaric, citric, methane sulfonic, formic, malonic, succinic, naphthalene-2-sulfonic, and benzene sulfonic. Also, pharmaceutically-acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.

The present invention additionally provides methods for treating or diagnosing a patient with therapeutic or diagnostic agents by delivering an effective amount of at least one therapeutic or diagnostic agent to cells for implementing the prevention, diagnosis, or treatment of a disease, condition, or syndrome, or symptoms thereof, which includes an alteration of the phosphorylation state of at least one enzyme and/or enzyme complex, or subunit thereof, including those characterized by cellular hyperproliferation.
Modulating the PDH complex as an improved treatment of cancer is especially contemplated, including treatment of primary tumors by the control of tumor cell proliferation, angiogenesis, metastatic growth, apoptosis, and treatment of the development of micrometastasis after or concurrent with surgical removal; and radiological or other chemotherapeutic treatment of a primary tumor. The pharmaceutical composition of the present invention is useful in such cancer types as primary or metastatic melanoma, lymphoma, sarcoma, lung cancer, liver cancer, Hodgkin's and non-Hodgkin's lymphoma, leukemias, uterine cancer, cervical cancer, bladder cancer, kidney cancer, colon cancer, and adenocarcinomas such as breast cancer, prostate cancer, ovarian cancer, and pancreatic cancer.

For therapeutic and diagnostic applications, the pharmaceutical composition can be administered directly to a patient when combined with a pharmaceutically-acceptable carrier.
This method may be practiced by administering the therapeutic or diagnostic agent alone or in combination with an effective amount of another therapeutic or diagnostic agent, which AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 AgTUffl0[TS/v(AW %.9888'' 0 may be, but is not limited to, a glycolytic inhibitor, a microtubule-interacting agent, a cytostatic agent, a folic acid inhibitor, an alkylating agent, a topoisomerase inhibitor, a tyrosine kinase inhibitor, podophyllotoxin or derivatives thereof, an antitumor antibiotic, a chemotherapeutic agent, an apoptosis-inducing agent, an anti-angiogenic agent, nitrogen mustards, nucleic acid intercalating agents, and combinations thereof. Such therapeutic agents may further include other metabolic inhibition reagents. Many such therapeutic agents are known in the art. The combination treatment method provides for simultaneous, sequential, or separate use in treating such conditions as needed to amplify or ensure patient response to the treatment method.

The methods of the present invention may be practiced using any mode of administration that is medically acceptable, and produces effective levels of the active compounds without causing clinically unacceptable adverse effects. Although formulations specifically suited for parenteral administration are preferred, the compositions of the present invention can also be formulated for inhalational, oral, topical, transdermal, nasal, ocular, pulmonary, rectal, transmucosal, intravenous, intramuscular, subcutaneous, intraperitoneal, intrathoracic, intrapleural, intrauterine, intratumoral, or infusion methodologies or administration, in the form of aerosols, sprays, powders, gels, lotions, creams, suppositories, ointments, and the like. If such a formulation is desired, other additives well-known in the art may be included to impart the desired consistency and other properties to the formulation.

Those skilled in the art will recognize that the particular mode of administering the therapeutic or diagnostic agent depends on the particular agent selected;
whether the administration is for treatment, diagnosis, or prevention of a disease, condition, syndrome, or symptoms thereof, the severity of the medical disorder being treated or diagnosed; and the dosage required for therapeutic efficacy. For example, a preferred mode of administering an anticancer agent for treatment of leukemia would involve intravenous administration, AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 ARMW ,Olltf/~0 10 AN0P
whereas preferred methods for treating skin cancer could involve topical or intradermal administration.

As used herein, "effective amount" refers to the dosage or multiple dosages of the therapeutic or diagnostic agent at which the desired therapeutic or diagnostic effect is achieved. Generally, an effective amount of the therapeutic or diagnostic agent may vary with the activity of the specific agent employed; the metabolic stability and length of action of that agent; the species, age, body weight, general health, dietary status, sex and diet of the subject; the mode and time of administration; rate of excretion; drug combination, if any; and extent of presentation and/or severity of the particular condition being treated. The precise dosage can be determined by an artisan of ordinary skill in the art without undue experimentation, in one or several administrations per day, to yield the desired results, and the dosage may be adjusted by the individual practitioner to achieve a desired therapeutic effect or in the event of any complication. Importantly, when used to treat cancer, the dosage amount of the therapeutic agent used should be sufficient to inhibit or kill tumor cells while leaving normal cells substantially unharmed.

The therapeutic or diagnostic agent included in the pharmaceutical compositions of the present invention can be prepared in any amount desired up to the maximum amount that can be administered safely to a patient. The amount of the diagnostic agent or therapeutic agent may range from less than 0.01 mg/mL to greater than 1000 mg/mL, preferably about 50 mg/mL.

Generally, the pharmaceutical composition of the present invention will be delivered in a manner sufficient to administer to the patient an amount effective to modulate the structure and/or activity of the PDH complex. The dosage amount may thus range from about 0.3 mg/rn 2 to 2000 mg/m2, preferably about 60 mg/m2. The dosage amount may be administered in a single dose or in the form of individual divided doses, such as from one to AMENDED SHEET - IPEA/US

PCTIUS08/04410 09-09-2008 P ll"J5 'QUS7 6Wi6%.D9:Zd08O10 four or more times per day. In the event that the response in a subject is insufficient at a certain dose, even higher doses (or effective higher doses by a different, more localized delivery route) may be employed to the extent of patient tolerance. Multiple doses per day are contemplated to achieve appropriate systemic or targeted levels of the therapeutic or diagnostic agent.

In yet another embodiment of the present invention, the lipoic acid derivatives of the present invention may be used as diagnostic and predictive agents in vitro. As stated earlier, depending on the specific tumor cell or cell type in question, different lipoic acid derivatives may be more or less effective at inhibiting distinct tumor classes through the modulation of the PDH complex. Thus, for example, in cases where diagnosis or selection of an appropriate chemotherapeutic strategy may be difficult, testing of a culture of tumor cells in vitro with lipoic acid derivatives known to target specific tumor cell types provides an alternative approach for identifying tumor types and effective treatments.

Turning to the figures, FIGURE 6A illustrates one of the many likely differences in energy metabolism in normal tissues and tumor cells in vivo. Tumor cells often rely more heavily on cytoplasmic glycolysis than mitochondrial energy metabolism for ATP
generation than do normal cells under corresponding conditions.. Changes in expression and regulation of the PDH complex are apparently part of this tumor-specific adaptation. The decreases in the levels of PDH catalytic components and/or increases in the levels of inhibitory PDKs producing these effects may render tumor cells much more vulnerable to agents attacking the PDH complex than are normal cells.

FIGURE 6B depicts the structures of lipoic acid as it catalyzes the normal reactions involved in synthesizing acetyl-CoA from pyruvate in the PDH complex. In vivo lipoic acid is joined through its carboxyl terminus in a non-peptide amide linkage to an epsilon amino group of a lysine in the E2 lipoyl domain active sites. Notice also that the AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 rP&TOOSWA InBA
$SS% %&p 10 oxidation/reduction/acetylation state of PDH E2-bound lipoate is monitored by the kinases and phosphatases that control PDH activity by controlling the phosphorylation inactivation of the Ela PDH subunit. This figure also depicts the structure of the three representative lipoic acid derivatives which may be used in the present invention. While CPI-613 and 5 have high anticancer potency, CPI-157 has little or no activity in cell culture and is useful as a control in several experiments.

FIGURE 6C presents the relationship between PDH complex components, including E2 with its bound lipoates, El, and the regulatory PDK. High levels of acetyl-lipoate or dihydrolipoate (not diagrammed) activate PDKs which, in turn, suppress further flux through 10 the PDH complex by inactivating Ela, the subunit catalyzing the first step in PDH complex catalysis. This process acts as a governor for carbon/energy flow through the PDH complex, and this regulatory process is apparently substantially altered to support the variant energy metabolism of tumor cells, as seen in FIGURE 3A.

FIGURE 7 shows the effects of the pharmaceutical composition of the present 15 invention on xenograft tumor growth. Cells were implanted subcutaneously on the dorsal flank as described in EXAMPLE 2. Mice were then injected with the drug (or vehicle alone;
"mock") intraperitoneally starting on days as indicated in the figure. The left panel shows a pancreatic tumor model injected three times weekly with the present invention at 1 mg/kg or the vehicle control. This experiment is representative of two done with BxPC-3 cells and of 20 two done with AsPC-1 cells. The right three panels show an H460 lung tumor model injected with the concentrations indicated either once weekly (circles), three times weekly (inverted triangles), or five times weekly (triangles for vehicle treatment and squares for drug treatment). This experiment is representative of four done with H460 cells.

FIGURE 8 shows the effect of treatment with the pharmaceutical composition of the 25 present invention on three tumor cell types and a non-transformed cell type (MDCK) either at A ffiNDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 FPCRUYZUU3700 14 N.09:Z OP 0 200-300 pM ("Treated") or mock treatment ("Mock treated"). Cells were treated in appropriate tissue culture media containing 10% serum or 48 hours. Extensive cell death by apoptosis or apoptosis-like pathways (see also FIGURE 11) in the three cancer cell lines is observed through the methodology described in EXAMPLE 2. In contrast, the non-transformed MDCK cells are apparently unaffected by drug treatment at this dose.

FIGURE 9A shows ATP levels in H460 lung cancer cells after treatment with the pharmaceutical composition of the present invention at or above the lethal threshold (200 M
in 10% serum). Dashed lines represent treatment at the concentrations indicated. Solid lines of corresponding texture represent treatment for the time indicated, followed by removal of the drug and 60 minutes of recovery in the drug-free medium. Block arrows indicated intervals of ATP recovery.

FIGURE 9B compares inhibition of ATP synthesis in media in which pyruvate (in the form of methyl-pyruvate) is the primary carbon source (dashed lines) and in which glucose is the primary carbon source (solid lines). Notice that the pharmaceutical composition of the present invention ultimately produces cell death at the same threshold concentrations in both media; however, early depletion of total cellular ATP levels is high in pyruvate-containing media and absent in glucose-containing media. Also, the onset of cell death is more rapid in the 300 M than the 200 M drug concentration.

FIGURE 9C compares the pharmaceutical composition of the present invention's inhibition of ATP synthesis in SK-Br-3 breast cancer cells and HMEC normal breast cells. In contrast to the experiments whose results are depicted in FIGURES 6A and 6B, these experiments were done in serum-free medium (MEBM). As a result, the drug's lethal threshold is lower, approximately 50 M. Note that the small depression in ATP
levels in the 22-hour normal cell samples is not related to drug dose and reflects normal experimental variation.

AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 O~-T 3 i~ $W-W.~6o~PI0 FIGURE 9D compares inhibition of ATP synthesis in H460 lung cancer cells by the pharmaceutical composition of the present invention (left graph), lipoic acid (center graph), and an inactive form of the present invention (right graph). As in FIGURE 9C, these experiments are done in serum-free medium so that the drug's lethal threshold is approximately 50pM.

FIGURE 10 illustrates the pharmaceutical composition of the present invention's effects (at 400 M in DMEM with 10% serum) on tumor cell mitochondria] levels of PDH
(PDC) and (aKDH) enzymatic activities. Notice that PDH is strongly inhibited whereas aKDH is not. Enzyme activity levels are measured in extracts of purified mitochondria using resazurin reduction in response to added carbon source, as described in EXAMPLE 2. The background line corresponds to resazurin reduction in the absence of added carbon source.
Next, in FIGURE 11 A, Western analyses of two-dimensional gels of extracts from H460 lung cancer cells treated (+) or mock treated (-) with the pharmaceutical composition of the present invention (at 400 M for 120 minutes in RPMI medium with 10% serum) were performed. The Western transfers were probed with a cocktail of monoclonal antibodies against Eta and E2 subunits of the PDH complex. The Western transfers are aligned at E2.
Notice the substantially higher levels of hyper-phosphorylated and the reduced levels of hypo-phosphorylated forms of El in the drug-treated sample. The left vertical white line illustrates one of the criteria for aligning the gels, the mobility of the E2 subunit. The right vertical white line passes through the less phosphorylated Eta form, the presumptively-enzymatically-active component.

FIGURE 11 B shows enlargements of paired two-dimensional gel samples treated and mock-treated with the pharmaceutical composition of the present invention.
Element A is an enlargement of a portion of FIGURE 8A. Element B is SK-Br-3 breast cancer cells mock-treated (-) and treated (+) for 180 minutes with 80 M of the composition in MEBM serum-AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 FF-TIMM t%.;U4&P10 free breast epithelial cell medium. Element C is SK-Br-3 breast cancer cells mock-treated (-) and treated (+) for 240 minutes with 80 M of the composition in MEBM serum-free breast epithelial cell medium. Element D is HMEC normal breast epithelial cells mock-treated (-) and treated (+) for 240 minutes with 80 M of the composition in MEBM serum-free breast epithelial cell medium. The vertical white line passes through the less phosphorylated Ela form, the presumptively-enzymatically-active component.

FIGURES 12A and 12B depict a working hypothesis for the strong, selective anticancer effects of the pharmaceutical composition of the present invention in vivo.
FIGURE 12A, for example, shows the regulatory role of PDKs as modulated by endogenous lipoate covalently bound to the PDC E2 subunit. PDKs normally inactivate the PDC in response to high levels of reduced and/or acetylated lipoate, a process that is apparently substantially modified in tumor cells.

Concomitantly, FIGURE 12B shows the large quantitative difference in the ratio of PDK to its substrate PDC-EI in the PDC, believed to distinguish normal and tumor cells in vivo. In normal cells the low level of PDK is thought to "walk" hand-over-hand (through its two dimeric subunits) around the PDH complex, gradually phosphorylating El.
This phosphorylation is in steady state equilibrium with PDP dephosphorylation (not diagrammed). On the working hypothesis diagrammed here, thioctans stimulate PDKs through the same sites that normally bind acetyl-lipoate and/or dihydrolipoate, thereby artificially stimulating one or more PDK isoform to inacative Ela. In cancer cells, the much higher levels of PDK might make this stimulation by thioctans much more effective in shutting down PDC enzymatic activity and mitochondrial energy metabolism.

The following non-limiting examples are provided to facilitate understanding of the pharmaceutical compositions of the present invention.

AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 pPCCf UUU7 7 f %A:9jaO, 0 CHEMICAL SYNTHESIS OF THIOCTANS

Lipoic acid derivatives (i.e., thioctans) CPI-613 and CPI-157 were synthesized by using a modified procedure described in US 6,331,559 BI and US 6,951,887 B2 with 6, 8-bismercaptooctanoic acid as the starting material. Thioctan CPI-045 was synthesized as described in US 6,331,559 B1.

Structure analyses for the three thioctans are below. Multiple independent syntheses of CPI-045 and CPI-613 were indistinguishable in their anti-cancer properties.
Purity of CPI-613 used in the xenograft (FIGURE 7) and ATP measurements (FIGURE 9) was in excess of 99%. All other preparations were greater than 98% pure.

CPI-613: 6,8-Bis-benzylsulfanyloctanoic acid: White crystalline solid, m.p. 65-(lit.' 67.5-69 ); 'H-NMR (250 MHz, CDCI3): 6 7.15-7.4 (m, 10H), 3.66 (s, 2H), 3.64 (s, 2H), 2.52-2.62 (m, 1H), 2.50 (t, J = 7.6 Hz, 2H), 2.29 (t, J = 7.6 Hz, 2H), 1.2-1.8 (m, 8H); 13C-NMR (62.9 MHz, CDC13): 8 179.6, 138.6, 138.5, 128.9, 128.8, 128.5, 128.4, 126.9, 44.1, 36.4, 35.1, 34.4, 33.8, 28.7, 26.0, 24.4.

CPI-157: 6,8-Bis-ethylsulfanyloctanoic acid: Colorless oil; TLC
(EtAc:Hexanes:HAc, 200:200:1 v/v): Rf = 0.60; 'H-NMR (300 MHz, CDC13): S 2.64-2.76 (m, 1H), 2.65 (t, J = 7.5 Hz, 2H), 2.52 (q, J = 7.5 Hz, 2H), 2.49 (q, J = 7.2 Hz, 2H), 2.36 (t, J
= 7.4 Hz, 2H), 1.40-1.85 (m, 8H), 1.25 (t, J = 7.2 Hz, 3H), 1.22 (t, J = 7.5 Hz, 3H); 13C-NMR

(75 MHz, CDC13): S 180.0, 44.3, 34.6, 33.9, 28.9, 26.2, 25.9, 24.5, 24.2, 14.9, 14.7; IR
(film): 2963, 1708, 1449, 1423, 1283, 1263 cm''.

CPI-045: 6,8-Bis-benzoylsulfanyloctanoic acid: Colorless, viscous oil; TLC
(Hexanes:EtAc:HAc, 100:50:1 v/v): Rf = 0.30; 'H-NMR (250 MHz, CDC13): S 7.9-8.1 (m, 4H), 7.38-7.60 (m, 6H), 3.8-4.0 (m, 1H), 3.0-3.3 (m, 2H), 2.34 (t, J = 7.1 Hz, 2H), 1.4-2.2 (m, 8H); 13C-NMR (62.9 MHz, CDC13): S 191.7, 191.5, 179.7, 137.0, 136.9, 133.3, 128.5, AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 ARM5~'v0'87vM0AN0V10 127.3, 127.1, 43.6, 35.0, 34.6, 33.8, 26.4, 26.2, 24.3; IR (film): 2973, 1710, 1704, 1667, 1665, 1662, 1448, 1207, 1175, 911, 773, 757, 733, 688, 648 cm Cells: Human tumor cell lines were obtained from ATCC and propagated according to ATCC recommendations. Human Mammary Epithelial Cells (HMEC), Small Airway Epithelial Cells (SAEC), and Normal Human Epidermal Keratinocytes (NHEK) primary cells were obtained from LONZA Walkersville, Inc (Walkersville, MD). Each cell line was 10 maintained and propagated in appropriate media developed by and bought from the supplier according to the supplier's instructions. Experiments reported here used normal cells at passage three to six.

Tumor growth inhibition studies: CD1-Nu/Nu female mice were implanted with human BxPC-3 or AsPC-1 pancreatic tumor cells or H460 NSCLC by subcutaneous (SC) 15 injection. Approximately 8-12 days later the mice were injected intraperitoneally (IP) at doses and schedule as indicated in the figure legend. Drug or vehicle was injected at ca. 2ml per 25gm of body weight. Drug concentration was 1.25mg/ml (ca. 3.1mM) or less.
The vehicle/solvent consisted of triethanolamine in water at 25mM or less. The vehicle injected in mock treated animals was always identical to the solvent in which the highest drug dose 20 for that experiment was injected. Mice were monitored daily for physical condition and mortality. Body weight and tumor volume were assessed daily before treatment, and approximately three times weekly during and after treatment. Mice were kept on a 12 hour light/dark cycle, were fed ad libitum and were housed at Stony Brook University Animal Facility in accordance with institutional guidelines.

25 Cell death assays: For most assessments of cell viability CellTiter-Glo assay AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 OFTTTW 642Q'If A9d O1 o (Promega) was used at times sufficiently long not to be confounded by early thioctan inhibition of ATP synthesis. (FIGURE 9). In a typical experiment, cells were plated in black, clear bottom, 96-well plates at 5,000 cells per well. 18-25 hours later, medium was replaced with fresh medium containing drug solvent (triethanolamine in water at 2.8mM in serum-containing media and 0.7mM in serum-free media) or thioctan CPI-613 in the same solvent. The assay was performed at 24 or 48 hours after drug addition, depending on drug dose, according the manufacturer's directions.

In some cases, cells were plated in 48-well plates at 10,000 cells per well, and medium was replaced 18-25 hours later with fresh medium containing drug solvent (-I%
EtOH final concentration) or different concentrations of thioctan CPI-045 in the same solvent. Cells remained in solvent or drug-containing medium for the remainder of the experiment. Plates were inspected at 24, 48 and 72 hours post drug addition, and cell numbers were estimated as a confluence percentage. Under these conditions, thioctan-induced cell death is highly apoptotic at near-threshold doses, and cell number estimates are very reliable indicators of death. (FIGURE 10) The integrity of cells remaining at 72 hours, if any, was tested by trypan blue exclusion.

Table 2 provides data regarding the action of thioctans against tumor cells in vitro.
Listed are the human tumor and primary human cells we have investigated for sensitivity to CPI-613 and/or CPI-045 killing. "+" indicates that the cells underwent apoptosis or necrosis-like cell death at doses of approximately 200-300 M (in the presence of 10%
serum) and approximately 50 pM in serum-free medium. (FIGURES 8 and 9) "--" indicates that these cells required approximately five-fold higher drug .doses to induce cell death in the corresponding medium. "nt" indicates non-tested combination. All tumor lines were analyzed in the appropriate media with 10% serum, as were the MDCK normal cells in FIGURE 8. In addition, HMEC, SAEC, NHKC primary human cells, and SK-BR-3, A549 AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 P&TuAZ 6%M' 7o.90 and H460 tumor lines were also analyzed in the appropriate serum-free media.
Primary cells were contact inhibited and transformed cells were at comparable densities.

ATP assay: Cells were plated in black, clear bottom, 96-well plates at 5,000 cells per well. 18-25 hours later, medium was replaced with fresh medium containing drug solvent (triethanolamine) or thioctan (CPI-613 or CPI-157) or lipoic acid for time interval and at drug concentration as indicated. Cell viability and integrity was assessed by recovery after drug withdrawal by trypan blue exclusion. ATP was measured using CellTiter-Glo luminescence assay (Promega) according to manufacturer's directions. All measurements were performed in duplicate and showed high consistency. The standard error of the mean ranged from 0.1-2% of the measured value. As a result, error bars were omitted from FIGURE 9.
Methyl pyruvate medium in FIGURE 9 consisted of RPMI without glucose (Invitrogen), supplemented with 10% dialyzed fetal bovine serum, 5mM HEPES (pH 7.4), and 10mM
methylpyruvate (Sigma-Aldrich), and the matched glucose medium was conventional RPMI
(Invitrogen).

PDH and aKDH enzyme assays: Tumor cells were grown to 80% confluence in 15 cm plates and treated with CPI-613 as indicated. Mitochondria were isolated according to the method of Moreadith and Fiskum. 1 Mitochondria were lysed in 0.4% lauryl maltoside. 50 l of mitochondrial lysate was added to 96-well plates. 50 ul of reaction mix (50mM Tris, pH
7.5, 2mM R-NAD+, 225uMv TPP, 2mM pyruvate or a-ketoglutarate, 150 M coenzyme A, 2.6mM cysteine, 1mM MgCl2) was added to mitochondrial lysates, and the mixture was incubated for 45 minutes at 37 C. At this time, 15 M resazurin and 0.5U/ml diaphorase were added to the mixture and incubated for an additional five minutes. NADH
production was monitored by measuring fluorescence using an excitation wavelength of 530nm and an emission wavelength of 590nm in a microplate reader (Fluorostar). All measurements were performed in duplicate and showed high consistency. The standard error of the mean ranged AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 ~c-T6Y UUU7H% 0 :OzJ:W(TBO' 0 from 0.3-4% of the measured value. As a result, error bars were omitted from FIGURE 10.
E 1 a phosphorylation:

Cell lysates for 2-D gels: Cells were grown to 95% confluence in 60 mm dishes and treated with drug or solvent as indicated. Cells were lysed in situ with 450 l Lysis buffer A
[455 l Zoom 2D protein solubilizer I (Invitrogen), 2.5 l 1M Tris base, 5.t1 IOOX protease inhibitor cocktail (Complete min, EDTA-free, Roche); 5 l 2M DTT]. Cell lysate was transferred to 1.5ml microfuge tubes and sonicated on ice for 15 passes at 50%
power. After minute incubation at room temperature, 2.5 l of dimethylacrylamide (DMA, Sigma-Aldrich) was added, and lysates were incubated for an additional 10 minutes. 5 t1 of 2M

10 DTT were added to neutralize excess DMA. Lysates were centrifuged at 16,000x g for 15 minutes.

2-D gels: We used Zoom Benchtop proteomics system (Invitrogen) according to the manufacturer's direction. Briefly, 30-50pl of lysate were mixed with 0.8111 pH

ampholytes, 0.75 1 2M DTT and brought up to 150 l with Zoom 2D protein solubilizer 1.

150 l of sample was loaded into 1PG runner, and pH 3-IONL IPG strips were added. Strips were soaked overnight at room temperature. A step protocol was used for isolectric focusing (250V, 20min.; 450V, 15min; 750V, 15 min 2000V, 30min). Strips were. treated for 15 minutes in 1X loading buffer, followed by 15 minutes in lX loading buffer plus 160mM
iodoacetatic acid. Strips were electrophoresed on NuPAGE 4-12% Bis Tris ZOOM
gels (Invitrogen).

AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 pPCT6W2 U81&gRI 19.D9:ZJOP

Table 2: Effect of thioctans against tumor and primary cells in vitro brain, glioblastoma U-87 MG + +
brain, glioblastoma LN-229 + nt breast, adenocarcinoma SK-Br-3 + +

breast, adenocarcinoma MCF7 nt +
bone, osteosarcoma Saos-2 nt +
cervical, adenocarcinoma HeLa + +
colorectal, adenocarcinoma SW480 nt +

hepatocellular, carcinoma Hep G2 + +
kidney, carcinoma A-498 + nt E lung, carcinoma A459 + +
F-lung, carcinoma H460 + +
muscle, rhabdomyosarcoma RD nt +
ovarian, carcinoma SKOV-3 + nt pancreatic, adenocarcinoma AsPC-1 + nt pancreatic, adenocarcinoma BxPC-3 + nt prostate, carcinoma LnCaP nt +
uterine, sarcoma Mes SA + +
uterine, sarcoma, MDR Mes-SA/dx5 + +

mammary epithelial cells HMEC -- --small airway epithelial cells SAEC -- nt a keratinocytes NHKC nt --AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 PP&TuWS
66%qj$%;J:QaogQ 10 zUW
Westerns: Proteins were blotted onto PVDF 4.5 m membranes. PDH Ela and E2 were detected using mAbs (Invitrogen).

Caspase-3 and PARP cleavage: Cleaved caspase-3 was detected on Western blots according to Roy and Nicholson.2 Briefly, after drug or solvent treatment cells were scraped 5 and the medium/cell/apoptotic bodies mixture was centrifuged at 6000x g.
Pellet was lysed with lysis buffer C (4M urea, 10% glycerol, 2% SDS, 0.003% BPB; 5% 2-mercaptoethanol).
30 g of total cell lysate protein per well were loaded on 12% Bis-Tris gels.
Proteins blotted onto PVDF 4.5 m membranes. Pro- and active-Caspase-3 were detected with anti-caspase-3 mAb (mouse monocolonal [31A1067]; abcam). PARP cleavage was detected using 10 monoclonal anti-poly (ADP-ribose) polymerase antibody, clone C-2-10 (Sigma-Aldrich).
Mitochondrial Ca+2 detection: Cells were seeded on 35 mm glass bottom plates (BD
Biosciences) at 3x105, grown overnight and treated with drug or solvent as indicated. Cells were then loaded with calcium dye Fluo-4, X-Rhod-1 or Rhod-2 (4 M, Invitrogen) in phenol red-free media and incubated at 37 C for 10 minutes. Cells were washed once with PBS, and 15 images were captured using an Axiovert 200M, (Zeiss) deconvolution microscope at a fixed exposure time, using FITC filter. Quantification of fluorescence was performed using software provided by the manufacturer. X-Rhod-1 and Rhod-2 gave similar results (FIGURE
13), indicating that these dyes were measuring a mitochondrial Ca +2 signal.34 References:
20 1. Moreadith RW and Fiskum G. Isolation of Mitochondria from Ascites Tumour-Cells Permeabilized with Digitonin. Analytical Biochemistry 137, 360-367 (1984).

2. Roy S and Nicholson DW. Criteria for identifying authentic caspase substrates during apoptosis. Apoptosis 322,110-125 (2000).

3. Gerencser AA and Adam-Vizi V. Selective, high-resolution fluorescence 25 imaging of mitochondria] Ca2+ concentration. Cell Calcium 30, 311-321 (2001).

AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 FP&74.90 4. Gyorgy H, Gyorgy C, Das S, Garcia-Perez C, Saotome M, Roy SS, and Yi MQ. Mitochondrial calcium signalling and cell death: Approaches for assessing the role of mitochondrial Ca2+ uptake in apoptosis. Cell Calcium 40, 553-560 (2006).

THIOCTANS PERTURB MITOCHONDRIAL MEMBRANE POTENTIAL AND Ca+2.
UPTAKE
The substrate effects on thioctan inhibition of ATP synthesis (FIGURE 9) indicate that the drug is interfering with the TCA cycle in the mitochondrial matrix.
If this is the case, we anticipate that mitochondrial membrane potential' might be compromised at lethal threshold doses and above. Using the potential-sensitive dye TMRE we observe the expected effect. (FIGURE 13) Mitochondrial membrane potential declines rapidly with initiation of drug treatment. The kinetics of membrane potential decline is very similar to the loss of ATP
synthesis in the presence of mitochondrial substrates. (FIGURE 9) ATP depletion in mitochondria is known to provoke a homeostatic response that includes the uptake of Ca+2 released from cytoplasmic stores, including the endoplasmic reticulum.2 Moreover, import of this Ca+2 into the mitochondrial matrix is thought to require the mitochondrial membrane potential. Thus, we anticipate that thioctan treatment at or above the lethal threshold may produce a sustained cytoplasmic release of Ca+2 with transient mitochondrial uptake of the ion in view of progressively compromised membrane potential.
Using X-Rhod-1 and Rhod-2 to measure mitochondrial Ca+2 and Fluo-4 to measure cytoplasmic Ca+2, we observe these expected effects. (FIGURE 13) By around two hours at CPI-doses at and slightly above the lethal threshold (compare FIGURES 9 and 13), as mitochondrial membrane potential declines, this initial mitochondrial Ca+2 transient decays. It is followed at 4-6 hours by a second large mitochondrial Ca +2 peak AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 $ %j.Z0&P s FF.T/ M 42 presumably associated with initiation of the calcium-dependent cell death pathways.3 References:

1. Garrett R and Grisham CM. Biochemistry. Thomson Brooks/Cole, Southbank, Vic., Australia; Belmont, CA. (2007).

2. Graier WF, Frieden M, and Malli R. Mitochondria and Ca2+ signaling: old guests, new functions. Pflugers Archiv-European Journal of Physiology 455, 375-(2007).

3. Gyorgy H, Gyorgy C, Das S, Garcia-Perez C, Saotome M, Roy SS, and Yi MQ. Mitochondrial calcium signalling and cell death: Approaches for assessing the role of mitochondrial Ca2+ uptake in apoptosis. Cell Calcium 40, 553-560 (2006).

THIOCTANS INDUCE DIVERSE CELL DEATH PROGRAMS

Reduction in mitochondrial energy metabolism is known to correlate with the decision to enter a cell death pathway in some circumstances, though detailed mechanisms remain incompletely understood.' "3 At thioctan doses above threshold but within -2-fold of this minimal killing dose, all tested cancer cell types undergo cell death that morphologically resembles apoptosis predominantly. (FIGURE 8) Apoptotic death was confirmed under these conditions by conventional Annexin immunostaning and TUNEL DNA end-labelling assays (results not shown).

At higher drug doses (more than -2-fold over threshold), active thioctans induce cell death (as assessed by replating viability assays and trypan blue or propidium exclusion) without the morphological correlates of apoptosis, suggesting a necrosis-like pathway (results not shown).

These data confirm that the thioctan CPI-613 inhibition mitochondrial energy AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 ARMOMId SANoP 0 metabolism correlates precisely with induction of cell death.

The observation that diverse tumor cells, known or presumed to contain inactivating mutations for distinct and diverse cell death pathways4 are all killed at very similar thioctan doses (FIGURE 8 and Table 2) is striking. This observation suggests that these drugs induce a master signal that is capable of engaging multiple, potentially redundant distal cell death execution pathways.5 Consistent with this possibility, we find that the Z-VAD-FMK generic caspase inhibitor subtly alters the morphology of cell death in thioctan treated cells, but has no discernible effect on the lethal threshold dose of the drug.

To further test the possibility that thioctan-induced cell death can proceed through multiple terminal execution mechanisms we examined caspase-3 and PARP-1 cleavage, diagnostic of distinct cell death pathways.5 We find that both thioctans CPI-613 and CPI-045 induce highly variable levels of these two cleavage events in different cells.
(FIGURE 14) Collectively, these results indicate that the thioctans are able to induce a strategic commitment to death that, depending on drug dose and cell type, is agnostic about the terminal, tactical execution of that decision.

References:
1. Watabe M and Nakaki T. ATP depletion does not account for apoptosis induced by inhibition of mitochondrial electron transport chain in human dopaminergic cells.
Neuropharmacology 52, 536-541 (2007).

2. Yuneva M, Zamboni N, Oefner P, Sachidanandam R, and Lazebnik Y.
Deficiency in glutamine but not glucose induces MYC-dependent apoptosis in human cells.
Journal of Cell Biology 178, 93-105 (2007).

3. Skulachev VP. Bioenergetic aspects of apoptosis, necrosis and mitoptosis.
Apoptosis 11, 473-485 (2006).

AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 FIFITU/Wagnp UII87Q0 W :0 NAO' O

4. Johnstone RW, Ruefli AA, and Lowe SW. Apoptosis: A link between cancer genetics and chemotherapy. Cell 108, 153-164 (2002).

5. Cregan SP, Dawson VL, and Slack RS. Role of AIF in caspase-dependent and caspase-independent cell death. Oncogene 23, 2785-2796 (2004).

LIPOIC ACID DERIVATIVE ANALOG STRUCTURES

Various non-limiting examples of lipoic acid derivative analogs presented below have been manufactured and are herein disclosed.

OH
S_s s.

61-r ~ N

OH
S's S, I \ I \
r__Y~ OH
.S S=S

OH
,S S

AMENDED SHEET - IPEA/US

PCT/US08/04410 09-09-2008 PPC9MW2 &6qR 0 0:9a0i 1 n--~j OH
crs /S
If ry--~j OH
S;piS

The foregoing discussion discloses and describes merely exemplary embodiments of 5 the present invention. One skilled in the art will readily recognize from such discussion, and from the accompanying claims, that various changes, modifications and variations can be made therein without departing from the spirit and scope of the invention as defined in the following claims. Furthermore, while exemplary embodiments have been expressed herein, others practiced in the art may be aware of other designs or uses of the present invention.

10 Thus, while the present invention has been described in connection with exemplary embodiments thereof, it will be understood that many modifications in both design and use will be apparent to those of ordinary skill in the art, and this application is intended to cover any adaptations or variations thereof. It is therefore manifestly intended that this invention be limited only by the claims and the equivalents thereof.

Claims (48)

1. A pharmaceutically-acceptable modulator of the regulation or perturbation of the structure, expression, and/or activity of at least one enzyme and/or enzyme complex, or subunit thereof, such as the modified pyruvate dehydrogenase (PDH) complex in the mitochondria of diseased cells of warm-blooded animals, including humans.
2. The modulator of claim 1, wherein the regulation or perturbation comprises reversible phosphorylation or dephosphorylation.
3. The modulator of claim 2, wherein the reversible phosphorylation or dephosphorylation occurs on a kinase, phosphatase, and/or dehydrogenase of an enzyme or enzyme complex, or subunit thereof.
4. The modulator of claim 3, wherein the modulator promotes or inhibits kinase activity.
5. The modulator of claim 4, wherein the kinase is selected from a group comprising pyruvate dehydrogenase kinase (PDK) 1, PDK2, PDK3, PDK4, and isoforms of each thereof.
6. The modulator of claim 3, wherein the modulator promotes or inhibits phosphatase activity.
7. The modulator of claim 6, wherein the phosphatase is selected from a group comprising pyruvate dehydrogenase phosphatase (PDP) 1, PDP2, and isoforms of each thereof.
8. The modulator of claim 3, wherein the modulator promotes or inhibits dehydrogenase activity.
9. The modulator of claim 2, wherein the reversible phosphorylation or dephosphorylation occurs at the PDH complex.
10. The modulator of claim 9, wherein the modulation occurs on the El a subunit of the PDH complex.
11. The modulator of claim 10, wherein the modulation occurs by inactivation of PDP and the isoforms and mutant forms thereof.
12. The modulator of claim 11, wherein the inactivation of PDP occurs by suppression of PDP expression.
13. The modulator of claim 10, wherein the modulation occurs by activation of PDK and the isoforms and mutant forms thereof.
14. The modulator of claim 1, wherein the diseased cells display sensitivity or insensitivity to treatment with the modulator of claim 1.
15. The modulator of claim 14, wherein treatment-insensitive diseased cells may be induced to express at least one modified enzyme or enzyme complex, or subunit thereof, so as to render them treatment-sensitive.
16. The modulator of claim 15, wherein expression is induced by genetic manipulation.
17. The modulator of claim 16, wherein the induction is achieved by transcriptional manipulation.
18. The modulator of claim 16, wherein the induction is achieved by translational manipulation.
19. The modulator of claim 16, wherein the induction is achieved by post-translational manipulation.
20. The modulator of claim 15, wherein expression is induced by epigenetic manipulation.
21. The modulator of claim 15, wherein expression is induced by phenotypic manipulation.
22. The modulator of claim 14, wherein the diseased cells express at least one modified enzyme or enzyme complex upon treatment with the modulator of claim 1.
23. The modulator of claim 1, wherein the modulator affects the expression level of PDK and the isoforms and mutant forms thereof.
24. The modulator of claim 1, wherein the modulator affects the expression level of PDP and the isoforms and mutant forms thereof.
25. The modulator of claim 23 or 24, wherein the expression level is altered at the level of transcription, translation, or post-translation.
26. The modulator of claim 25, wherein the alteration is epigenetic.
27. The modulator of claim 9, wherein the modulator inhibits the creation of toxic metabolites.
28. The modulator of claim 9, wherein the modulator promotes the detoxification of toxic metabolites.
29. The modulator of claims 27 or 28, wherein the metabolites are selected from a group consisting of acetaldehyde, superoxide, hydrogen peroxide, and hydroxyl radical.
30. The modulator of claim 28, wherein the effect of modulation is observed by a decrease in acetoin production.
31. The modulator of claim 9, wherein the reversible phosphorylation or dephosphorylation becomes irreversible.
32. The modulator of claim 31, wherein the effect of the phosphorylation or dephosphorylation results in cell death.
33. The modulator of claim 32, wherein the effect is apoptosis.
34. The modulator of claim 32, wherein the effect is necrosis.
35. The modulator of claim 1, comprising at least one lipoic acid derivative and at least one pharmaceutically-acceptable carrier thereof.
36. The modulator of claim 35, wherein the lipoic acid derivative has the formula:

wherein R1 and R2 are independently selected from the group consisting of hydrogen, alkyl Cn,H2n+1, alkene CnH2n, alkenyl CnH2n-1, alkyne CnH2n-2, alkynyl CnH2n-3, alkyl sulfide CH3(CH2)n-S-, disulfide alkyl CH3CHt-S-S-, thiocarbamic ester (CH2)nC=NH-, and semithioacetal CH3CH(OH)-S-, wherein n is 1-10 and t is 0-9; aromatic; acyl defined as R3C(O)-; heteroaryl; imidoyl defined as R4C(=NH)-; organometallic aryl; alkyl-organometallic aryl; and semiacetal R5CH(OH)-S-;

wherein R1 and R2 as defined above can be unsubstituted or substituted;

wherein R3 is selected from the group consisting of hydrogen, alkenyl, alkynyl, alkylaryl, heteroaryl, alkylheteroaryl and organometallic aryl, any of which can be substituted or unsubstituted;

wherein R4 is selected from the group consisting of hydrogen, alkenyl, alkynyl, aryl, alkylaryl, heteroaryl, and alkylheteroaryl, any of which can be substituted or unsubstituted;
wherein R5 is CC13, CF3, or COOH;

and wherein x is 0-16;
metabolites thereof;
or salts thereof.
37. The modulator of claim 35, wherein the lipoic acid derivative has the formula:

wherein M is a covalent bond, -[C(R1)(R2)]Z , or a metal chelate or other metal complex where the metal is not palladium;

wherein R1 and R2 are independently selected from the group consisting of hydrogen, acyl R3C(O)-, alkyl CnH2n+1, alkenyl defined as CnH2n-1, alkynyl defined as CnH2n-3, aryl, heteroaryl, alkyl sulfide CH3(CH2)n-S-, imidoyl defined as R3C(=NH)-, and hemiacetal defined as R4CH(OH)-S-;

wherein R1 and R2 as defined above can be unsubstituted or substituted;

wherein R3 and R4 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, alkylaryl, heteroaryl, and heterocyclyl, any of which can be substituted or unsubstituted;

wherein R5 is selected from the group consisting of -CC13, -CF3 or -COOH;
and wherein x is 0-16, z is 0-5, and n is 0-10;

metabolites thereof;
or salts thereof.
38. The modulator of claim 35, wherein the lipoic acid derivative has the formula:

wherein R1 and R2 are independently selected from the group consisting of hydrogen, alkyl CnH2n+1, alkene CnH2n, alkenyl CnH2n-1, alkyne CnH2n-2, alkynyl CnH2n-3, alkyl sulfide CH3(CH2)n-S-, disulfide alkyl CH3CHt-S-S-, thiocarbamic ester (CH2)nC=NH-, and semithioacetal CH3CH(OH)-S-, wherein n is 1-10 and t is 0-9, aromatic, acyl defined as R4C(O)-, heteroaryl, imidoyl defined as R5C(=NH)-, organometallic aryl, alkyl-organometallic aryl, semiacetal R6CH(OH)-S-, amino acids, carbohydrates, nucleic acids, lipids, and multimers and combinations thereof;

wherein R1 and R2 can be unsubstituted or substituted;

wherein R3 is selected from a group consisting of amino acids, carbohydrates, nucleic acids, lipids, and multimers thereof;

wherein R4 is selected from the group consisting of hydrogen, alkenyl, alkynyl, alkylaryl, heteroaryl, alkylheteroaryl and organometallic aryl, any of which can be substituted or unsubstituted;

wherein R5 is selected from the group consisting of hydrogen, alkenyl, alkynyl, aryl, alkylaryl, heteroaryl, and alkyiheteroaryl, any of which can be substituted or unsubstituted;
wherein R6 is CC13, CF3, or COOH;

and wherein x is 0-16;
metabolites thereof;
or salts thereof.
39. The modulator of claim 35, wherein the lipoic acid derivative has the formula:

wherein M is a covalent bond, -[C(R1)(R2)]Z , or a metal chelate or other metal complex where the metal is not palladium;

wherein R1 and R2 are independently selected from the group consisting of hydrogen, acyl R4C(O)-, alkyl CnH2n+1, alkenyl defined as CmH2m-1, alkynyl defined as CmH2m-3, aryl, heteroaryl, alkyl sulfide CH3(CH2)n-S-, imidoyl defined as R4C(=NH)-, hemiacetal defined as R6CH(OH)-S-, amino acids, carbohydrates, nucleic acids, lipids, and multimers and combinations thereof;

wherein R1 and R2 can be unsubstituted or substituted;

wherein R3 is selected from a group consisting of amino acids, carbohydrates, nucleic acids, lipids, and multimers thereof;

wherein R4 and R5 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, alkylaryl, heteroaryl, and heterocyclyl, any of which can be substituted or unsubstituted;

wherein R5 is selected from the group consisting of CCl3, CF3 or COOH;
and wherein x is 0-16, z is 0-5, n is 0-10 and m is 2-10;

metabolites thereof;
or salts thereof.
40. The modulator of claims 36, 37, 38, or 39, wherein the lipoic acid derivative is present solely as the (R)-isomer thereof.
41. The modulator of claims 36, 37, 38, or 39, wherein the lipoic acid derivative is present as a mixture of the (R)-isomer and the (S)-isomer thereof.
42. The modulator of claim 1, wherein the modulator is useful in the treatment and diagnosis of a disease, condition, or syndrome, or symptoms thereof, which includes an alteration of the structure, expression, and/or activity of at least one enzyme and/or enzyme complex, or subunit thereof.
43. The modulator of claim 42, wherein the at least one enzyme complex is the PDH complex.
44. The modulator of claim 42, wherein the disease, condition, or syndrome is further characterized by cellular hyperproliferation.
45. The modulator of claim 44, wherein the disease, condition, or syndrome is cancer.
46. A method of modulating at least one enzyme and/or enzyme complex, or subunit thereof, in a patient presenting a disease, condition, or syndrome which includes an alteration of the structure, expression, and/or activity of the at least one enzyme and/or enzyme complex, or subunit thereof, comprising administration of an effective amount of the modulator of claim 1.
47. The method of claim 46, wherein at least one enzyme complex is the PDH
complex.
48. The method of claim 46, wherein the disease, condition, or syndrome is further characterized by cellular hyperproliferation.

50. The method of claim 48, wherein the disease, condition, or syndrome is cancer.

51. A method of diagnosing and predicting benefit in a patient presenting symptoms of a disease, condition, or syndrome which includes an alteration of the structure, expression, and/or activity of at least one enzyme and/or enzyme complex, or subunit thereof, comprising obtaining a sample of cells from the patient, administering an effective amount of the modulator of claim 1 to the cells in vitro, and obtaining the results therefrom.

52. The method of claim 51, wherein at least one enzyme complex is the PDH
complex.

53. The method of claim 51, wherein the disease, condition, or syndrome is further characterized by cellular hyperproliferation.

54. The method of claim 53, wherein the disease, condition, or syndrome is cancer.
CA2720396A 2008-04-04 2008-04-04 Pharmaceutical composition Abandoned CA2720396A1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2008/004410 WO2009123597A1 (en) 2008-04-04 2008-04-04 Pharmaceutical composition

Publications (1)

Publication Number Publication Date
CA2720396A1 true CA2720396A1 (en) 2009-10-08

Family

ID=41135840

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2720396A Abandoned CA2720396A1 (en) 2008-04-04 2008-04-04 Pharmaceutical composition

Country Status (10)

Country Link
EP (1) EP2268278A4 (en)
JP (1) JP2011516473A (en)
KR (1) KR20110025168A (en)
CN (1) CN102056605A (en)
AU (1) AU2008354009A1 (en)
BR (1) BRPI0821894A2 (en)
CA (1) CA2720396A1 (en)
IL (1) IL208387A0 (en)
MX (1) MX2010010867A (en)
WO (1) WO2009123597A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX343847B (en) 2010-07-08 2016-11-24 Wrigley W M Jun Co Chewing gum containing a cross-linked gelatin matrix gum base.
US9421234B2 (en) * 2011-05-20 2016-08-23 Nutramax Laboratories, Inc. Orally administrable compositions comprising avocado/soybean unsaponifiables and lipoic acid and methods of administration
CA2863600A1 (en) * 2012-02-16 2013-08-22 The Penn State Research Foundation Modulators of acyl-coa lysocardiolipin acyltransferase 1 ( alcat1) and uses thereof
CN105142643B (en) * 2012-12-19 2021-09-03 基石医药公司 Pharmaceutical compounds
AU2014398232B2 (en) 2014-06-19 2019-11-28 Rafael Pharmaceuticals, Inc. Pharmaceutical compounds
WO2015195071A1 (en) 2014-06-19 2015-12-23 Robert Shorr Pharmaceutical compounds
CN110590624B (en) * 2019-09-26 2021-02-26 苏州富士莱医药股份有限公司 Preparation method of lipoic acid derivative 8- (ethyl disulfide) -6- (phenyl disulfide) caprylic acid
WO2023012305A1 (en) * 2021-08-04 2023-02-09 Cytacoat Ab Disulfide compounds as medicaments

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2853497A (en) * 1953-12-04 1958-09-23 Merck & Co Inc 6, 8-bis (hydrocarbon substituted mercapto) 5-hydroxycaprylic acids and delta-lactones thereof
US2789991A (en) * 1954-05-20 1957-04-23 American Cyanamid Co Substituted disulfones
JPS61198148A (en) * 1985-01-29 1986-09-02 Fuji Photo Film Co Ltd Silver halide photographic sensitive material
GB9618934D0 (en) * 1996-09-11 1996-10-23 Univ London Inositol phosphoglycans for therapeutic use in the treatment of diabetes and obesity
DK1624060T3 (en) * 1998-03-20 2012-04-10 Commw Scient Ind Res Org Control of gene expression
FR2777001B1 (en) * 1998-04-01 2000-06-09 Cird Galderma 6,8-DIMERCAPTOOCTANOIC ACID DERIVATIVES SUBSTITUTED IN 6-S AND / OR 8-S BY RADICAL (3-METHYLTHIOPROPANOYL) AND PHARMACEUTICAL COMPOSITIONS FOR THE TREATMENT OF CANCER TUMORS
US6331559B1 (en) * 1998-10-26 2001-12-18 The Research Foundation Of State University Of New York At Stony Brook Lipoic acid derivatives and their use in treatment of disease
DE19904794A1 (en) * 1999-02-05 2000-08-10 Biotechnolog Forschung Gmbh Methods for improving the primary metabolism of mammalian cell lines
US6284786B1 (en) * 1999-02-16 2001-09-04 The Center For The Improvement Of Human Functioning, Int'l., Inc. Treatment of cancer using lipoic acid in combination with ascorbic acid
AR042572A1 (en) * 1999-04-02 2005-06-29 Sod Conseils Rech Applic DERIVATIVES OF LIPOIC ACID, PROCEDURE FOR THE PREPARATION, MEDICINES AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM AND USE OF SUCH DERIVATIVES FOR THE PREPARATION OF THE REFERRED MEDICATIONS
IT1312060B1 (en) * 1999-04-09 2002-04-04 Antibioticos Spa USE OF ALPHA LIPOIC ACID IN ANTIMETASTATIC TREATMENT.
US7108977B2 (en) * 2000-05-19 2006-09-19 Hoffmann-La Roche Inc. Process for determining the tumoricidal potential of a sample the use of a nucleic acid which is downregulated in human tumor cells
WO2006119355A2 (en) * 2005-05-03 2006-11-09 Albert Einstein College Of Medicine Of Yeshiva University Mammalian hypothalamic nutrient modulation of glucose metabolism
US20070077583A1 (en) * 2005-09-21 2007-04-05 Aurelium Biopharma, Inc. Alpha enolase-directed diagnostics and therapeutics for cancer and chemotherapeutic drug resistance
US20070212360A1 (en) * 2006-01-17 2007-09-13 Denko Nicholas C Modulation of mitochondrial oxygen consumption for therapeutic purposes
JP5412423B2 (en) * 2007-04-18 2014-02-12 コーナーストーン ファーマシューティカルズ,インコーポレーテッド Lipoic acid derivative
AU2008242827B2 (en) * 2007-04-18 2014-06-05 Cornerstone Pharmaceuticals, Inc. Pharmaceutical formulations containing lipoic acid derivatives
BRPI0821176A2 (en) * 2008-03-04 2015-10-06 Robert Rodriguez phosphorylation state modulator of at least one enzyme complex, method for modulating the pdh complex in a patient presenting a disease, condition, or syndrome, and method for diagnosing and predicting benefit in a patient presenting symptoms of a disease, condition , or syndrome caused by a change in the structure and / or activity of the pdh complex

Also Published As

Publication number Publication date
EP2268278A4 (en) 2011-11-09
BRPI0821894A2 (en) 2015-07-21
AU2008354009A1 (en) 2009-10-08
WO2009123597A1 (en) 2009-10-08
EP2268278A1 (en) 2011-01-05
JP2011516473A (en) 2011-05-26
CN102056605A (en) 2011-05-11
MX2010010867A (en) 2011-05-25
IL208387A0 (en) 2010-12-30
KR20110025168A (en) 2011-03-09

Similar Documents

Publication Publication Date Title
US8552050B2 (en) Activators of pyruvate kinase M2 and methods of treating disease
CA2720396A1 (en) Pharmaceutical composition
ES2663789T3 (en) Composed of pyrazole-amide and medicinal uses thereof
Gan et al. Identification of Michael acceptor-centric pharmacophores with substituents that yield strong thioredoxin reductase inhibitory character correlated to antiproliferative activity
US9642874B2 (en) Compositions of selenoorganic compounds and methods of use thereof
CA2638735A1 (en) Methods to identify inhibitors of the unfolded protein response
Jin et al. Design, synthesis and activity of Mnk1 and Mnk2 selective inhibitors containing thieno [2, 3-d] pyrimidine scaffold
Lougiakis et al. Synthesis and pharmacological evaluation of novel adenine–hydrogen sulfide slow release hybrids designed as multitarget cardioprotective agents
CA2717511A1 (en) Modulation of enzymatic structure, activity, and/or expression level
CA2739250C (en) Selective proton coupled folate transporter and folate receptor, and garftase inhibitor compounds and methods of using the same
Li et al. Discovery of novel exceptionally potent and orally active c-MET PROTACs for the treatment of tumors with MET alterations
EP3189033B1 (en) Pharmaceutical compounds
US20140243401A1 (en) Methods of screening compounds that are cytotoxic to tumor cells and methods of treating tumor cells using such compound
Oh et al. Selective targeting of cancer cells using a hydrogen peroxide-activated Hsp90 inhibitor
CA2965898C (en) Fatty acid analogs
CA2895029A1 (en) Metal complexes of lipoic acid derivatives
US11773096B2 (en) Small-molecule PI5P4K alpha/beta inhibitors and methods of treatment using same
Hatzivassiliou et al. Akt-directed metabolic alterations in cancer
Zhou et al. Structural Optimization of Marine Natural Product Pretrichodermamide B for the Treatment of Colon Cancer by Targeting the JAK/STAT3 Signaling Pathway
Hu Development of Membrane-Protein Interaction Modulators as Allosteric Drug Candidates
Lengauer Targeting glutamine metabolism and mitochondria via natural compounds in cancer: novel therapeutic strategies
KR101384349B1 (en) Anti-Cancer Composition Comprising An Inhibitor for Serine-Palmitoyltransferase As Active Ingredient

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20130403

FZDE Discontinued

Effective date: 20151008