Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Feb 1.
Published in final edited form as: Curr Opin Chem Biol. 2022 Dec 12;72:102235. doi: 10.1016/j.cbpa.2022.102235

Mitochondria as a target of third row transition metal-based anticancer complexes

Chibuzor Olelewe 1, Samuel G Awuah 1,2,3,*
PMCID: PMC9870944  NIHMSID: NIHMS1851446  PMID: 36516614

Abstract

In pursuit of better treatment options for malignant tumors, metal-based complexes continue to show promise as attractive chemotherapeutics due to tunability, novel mechanisms, and potency exemplified by platinum agents. The metabolic character of tumors renders the mitochondria and other metabolism pathways fruitful targets for medicinal inorganic chemistry. Cumulative understanding of the role of mitochondria in tumorigenesis has ignited research in mitochondrial targeting metal-based complexes to overcome resistance and inhibit tumor growth with high potency and selectivity. Here, we discuss recent progress made in third row transition metal-based mitochondrial targeting agents with the goal of stimulating an active field of research toward new clinical anticancer agents and the elucidation of novel mechanisms of action.

1.0. Introduction

Mitochondria are organelles prominent for their major function, which is ATP production[1]. However, the mitochondria also play a major role in many important cellular processes including anabolic (synthesis of biomolecules) and catabolic processes, regulated cell death processes, cell signaling, and redox homeostasis[1,2]. For many years, following Otto Warburg’s discovery of aerobic glycolysis in cancer cells[3], it was widely accepted that the mitochondria are defective in cancer cells despite Weinhouse’s seminal report on active mitochondria in tumors[4]. Increasing extensive investigation of mitochondria processes in cancer over the past decades have highlighted the crucial role of functional mitochondria in cancer cell growth and survival. Thus, creating impetus to develop strategies aimed at targeting the mitochondria for effective cancer treatment[58]. The field of medicinal inorganic chemistry has seen significant advancement since the discovery of the anticancer activity of the platinum-based metal complex, cisplatin[911]. The success of cisplatin and its analogs in the clinic has been hampered by adverse effects in patients[12,13], acquired tumor resistance, and recurrence in many aggressive tumors including lung, bladder, ovarian, and breast tumors. Therefore, efficacious metal-based complexes with high potency and enhanced selectivity for tumor cells over normal cells remain an unmet need. Metabolic heterogeneity and prominent dependence of some aggressive cancers such as PDAC, and glioblastoma on mitochondrial metabolism present liabilities that can be exploited by mitochondrial targeting agents. The redox active character of transition metals provides opportunities for optimization to target the inherently active mitochondria organelle. Additionally, mitochondria-targeted metallodrugs generally exhibit a differentiated cytotoxic mechanism of action from the FDA approved platinum drugs, with implications to circumvent the problem of tumor resistance associated with platinum drugs. To this end, several novel platinum and non-platinum complexes such as gold[14,15], ruthenium[1618], rhenium[19], iridium[2022] osmium[23] complexes with mitochondria targeting ability have been developed over the years. In this review, we provide an up-to-date development of metallo-agents targeting the mitochondria with translational potential and with primary focus on third-row transition metals (Figure 1). We regret to have omitted closely related work in the field due to limited word constraints for this review.

Figure 1.

Figure 1.

Summary of mechanism-of-action of selected metal-based complexes in this review.

2.0. Mitochondria-targeted platinum complexes

The impact of the FDA approved platinum drugs for cancer treatment has been revolutionary. To overcome limiting side effects such as drug resistance and debilitating side effects, different structural scaffolds that include monofunctional Pt(II)[24,25] and dual-threat Pt(IV) have been developed[26,27]. Whereas pyriplatin and phenanthriplatin bind DNA nucleobases in a monodentate fashion, chemical modification of pyriplatin to incorporate delocalized positive charges in triphenylphosphonium (Ph3P+, TPP) moieties on the pyridine coordinating ligand (OPT, MPT, PPT, TTP, Mon-Pt) leads to inhibition of mitochondrial bioenergetics, mtDNA, and the perturbation of glucose metabolism[28] (Figure 2). Similarly, as with mitaplatin[29], and Platin-M[30], the axial functionalization of cisplatin with TPP, dichloroacetates, luminogens, (PMT[31], DPB[26], and ACPt[32]), etcetera to form Pt(IV) prodrugs facilitates mitochondria delivery and subsequent modulation of oxidative phosphorylation (OXPHOS) and mtDNA inhibition towards cell death[26,32]. The ability of these agents to inhibit Pt-resistant cells and tumor growth in vivo demonstrates their clinical relevance.

Figure 2.

Figure 2.

Chemical structures of mitochondria targeting platinum complexes.

3.0. Mitochondria-targeted gold complexes

Gold-based therapeutic agents have emerged among the leading class of metal-based drugs that are being developed as potential alternatives to platinum-based drugs[14,3335]. This is energized by the several repurposing and current phase II clinical trials of the FDA approved gold drug, auranofin in ovarian and non-small cell lung cancer. Different structural variations of gold-based complexes exist, including cyclometalated gold complexes, gold thiolates, gold porphyrins, gold NHCs, and gold-phosphine complexes[14]. Although their molecular mechanism of action is a subject of intense investigation, the mitochondrion is implicated as a potential target responsible for the observed cytotoxic effect of some of these complexes[3640]. Conceivably, cationic gold complexes with lipophilic character are susceptible to mitochondrial localization due to the negative mitochondrial membrane potential in cancer cells that drive selectivity. Recent target identification strategies such as chemical proteomics, thermal proteome profiling, and click chemistry have accelerated the identification of novel targets including mitochondrial proteins in metallodrug discovery. Early work by Berners-Price uncovered mitochondria-targeting gold chemotherapeutics[15,41,42]. Che and colleagues reported the development of gold(III) complexes that interact with mitochondrial proteins. Notably, a mesoporphyrin complex (AuMesoIX) exerts its anticancer activity via a unique mechanism that involves gold(III) ion activation of the porphyrin ring towards adduct formation with cysteine thiols found in proteins important for cancer survival including mitochondrial peroxiredoxin (PRDX3) and HSP90, thioredoxin (TrxR) and protein deubiquitinases (DUBs) (Figure 3a)[43]. AuMesoIX inhibits cancer cell growth in the nanomolar and submicromolar range, distorts mitochondria morphology, and inhibits tumor growth in ovarian and lung xenograft mouse models.

Figure 3.

Figure 3.

Chemical structures of mitochondria targeting gold complexes.

Cyclometalated gold complexes have become an attractive structural class for anticancer action due to the strong sigma donation that impart enhanced stability [36,40,4446]. In addition to electrochemical stability often derived from cyclometalation, the strong Au-C covalent bond character promotes solution stability even under physiological conditions to overcome premature deactivation[14]. Recently, the development of cyclometalated Au(III) complexes bearing metformin or phenformin biguanides (Figure 3c) [40,47] demonstrates significant anticancer potential in vitro and in vivo by altering mitochondrial bioenergetics. Specifically, Auraformin significantly accumulates in the mitochondria and efficiently impair mitochondria respiration in triple negative breast cancer (TNBC) MDA-MB-468 cells as well as depolarize the mitochondria membrane in the cells and 3met severely disrupts mitochondria metabolism in MDA-MB-231 cells leading to ER stress and autophagy. 3met demonstrates in vivo efficacy in a MDA-MB-231 orthotopic xenograft mouse model.

Analogs of a unique cyclometalated gold(III) complex bearing bisphosphine ligands known as the AuPhos class of compounds interfere with the mitochondrial electron transport chain (ETC) and mitochondria biogenesis [36,37]. Lead complexes, AuPhos-89 and AuPhos-19 (Figure 3b and 3d) display potent inhibitory effects in a panel of cancer cell lines including the recalcitrant TNBC cell lines and inhibits tumor growth in syngeneic and xenograft models of TNBC in mice. The novel mechanism of AuPhos-89 to promote mitochondria biogenesis has facilitated its use in the treatment of the inflammatory disease, ulcerative colitis[4850].

The Awuah lab developed new tricoordinate gold(I) complexes (AuTri-9) (Figure 3f) capable of disrupting mitochondrial structure. In addition to transmission emission microscopy images and quantification of cristae length, immunoblotting showed depletion of key mitochondrial fusion and fission proteins such as OPA1, MFN, MFF and TOM20 which are involved in the maintenance of mitochondrial structure and dynamics[51]. Taken together, ligand tuning at the gold center and oxidation state contribute to modulating distinct pathways of the mitochondria for therapeutic gain. We posit that this exciting area of research will generate tool compounds to study mitochondrial metabolism towards uncovering elusive targets and develop therapeutic agents to treat metabolic and non-metabolic diseases.

4.0. Mitochondria-targeted iridium complexes

Third row iridium-based complexes possess peculiar triplet excited state characteristics that make them useful for phosphorescence applications including bioimaging and photosensitization, especially in photodynamic therapy (PDT). Organelle selective iridium complexes that interact with the ER, lysosome, and the mitochondria hold tremendous promise as anticancer agents than conventional treatments [5257]. The development of cell-permeable cyclometalated Ir(III) complexes as photosensitizers for PDT is attractive due to their efficient singlet oxygen (1O2) production required for tumor damage and luminescent character for theranostics (Figure 4a and 4c) [58,59]. Furthermore, the potential for structural optimization via rational ligand design and tuning provides opportunities to expand the diversity of iridium-based complexes to achieve more photostable and better phototoxic complexes. Recent work by Zhuezhao et. al. described the development of new dinuclear IrIII-based metallohelices that target mitochondria DNA with enhanced PDT tunability in cancer cells[58]. The synthesis of these IrIII-based triple stranded metallohelices was achieved by reacting the aldehyde functionalized fac-Ir(ppy)3 (ppy=2-phenylpyridine) and linear alkanediamine spacers via imine-coupling to afford stereochemically distinct helicates (H4b and H2b) or mesocates (M5b and M3b) (Figure 4a). These structures localize predominantly in the mitochondria, interact with mitochondria DNA, and generate cytotoxic 1O2 upon irradiation. This work highlights the potential for mitochondria-DNA targeted IrIII-based metallohelices for PDT.

Figure 4:

Figure 4:

Chemical structures of mitochondria targeting iridium complexes. (a) Ir(III)-based metallohelices. Reproduced with permission from ref.[58]. Copyright 2020 John Wiley and Sons. (b) Analogs of Ir complexes targeting mitochondrial topoisomerase. (c) Iridium (III) based complex for photodynamic therapy/photoactivated therapy. Reproduced with permission from ref.[59]. Copyright 2022 American Chemical Society. (d) Chemical structures of mitochondria targeting rhenium complexes.

Phototoxic octahedral Ir(III) complexes undergo Type I photochemical mechanism under hypoxic conditions to induce cancer cell death in 2D culture and 3D spheroids[60]. A derivative of the cationic Ir(III) thienyl benzimidazole of the formula [Ir(C^N)2(dppz)][PF6], where C^N = 1-methyl-2-(2′-thienyl)benzimidazole, significantly inhibits the growth of the aggressive rhabdomyosarcoma (RD) cells, a rare form of pediatric cancer and cancer stem cells[61]. The complex accumulates in the mitochondria and induces depolarization of the mitochondria membrane potential (MMP), which may be a premise for the selective cancer stem cell targeting reported[62]. Despite the promising effect of the complex in bulk and CSCs, more work is required to delineate the mechanism of action, which implicates the mitochondria.

Beyond PDT, Liting et. al. developed two iridium complexes (Ir1 and Ir2) that target mitochondria topoisomerase (Figure 4b) [63]. The concentration dependent inhibitory effect of the Ir(III) complex on mitochondrial topoisomerase I (TOP1mt) is comparative to the clinically used anticancer drug, camptothecin (CPT). The synthetic design of these complexes leverages the topoisomerase I inhibiting ability of indenoisoquinoline moieties and the mitochondria-targeting Ir(III) core to develop novel mitochondrial topoisomerase I targeting Ir(III) complexes. These complexes demonstrate high efficacy against cancer cells (40-fold > than cisplatin) with no cross-resistance in cisplatin resistant cells (A549R).

5.0. Mitochondria-targeted Rhenium complexes

The upward trajectory in the development of rhenium organometallics continues with recent reports of new potent anticancer rhenium complexes highlighting the progress made[6468]. The design of mitochondria targeting rhenium(I) core to incorporate the FDA approved iron chelator, deferasirox affords the complex, DFX-Re3, which is capable of relocating intracellular iron into the mitochondria and disrupt mitochondria metabolism (Figure 4d)[67]. Acting at the interface between mitochondria process and epigenetic modification, DFX-Re3 transfer of iron downregulates Fe(II)/ α-ketoglutarate -dependent demethylases and causes a marked increase in DNA, RNA and histone methylation. DFX-Re3 modulates levels of metabolites such as α-ketoglutarate, succinate, and fumarate associated with epigenetic regulation. Moreover, DFX-Re3 inhibit TNBC tumor growth in 4T1 tumor-bearing mice at a dose of 5 mg/kg over a 14-day treatment period. Given that there was no observed toxicity on organs after the treatment duration, it ignites enthusiasm in overcoming deferasirox-associated nephrotoxicity and the advancement of this complex for TNBC chemotherapy[69].

Dinuclear rhenium(I) tricarbonyl complexes linked via a diazo (ReN) or disulfide (ReS) bridge targets the mitochondria and causes oxidative stress induced cell death in cancer cells (Figure 4d) [68]. The complexes display cytotoxic action against different cancer cell lines including the cisplatin resistant A549R. ReN and ReS inhibit mitochondria metabolism and decreased mtDNA copy number. An examination of the effect of these complexes on the gene expression profile showed downregulation of 13 mtDNA encoded genes after treatment with the complexes. The encouraging in vivo efficacy of these complexes with minimal toxicity highlights the excitement for rhenium organometallics. Other Re(I) complexes (Re1)[70] including those bearing artesunate ligands (Re-ART-1 and Re-ART-2) (Figure 4d)[71] have been shown to modulate mitochondria function. Opportunities remain for the development of mitochondrial targeting Re complexes.

6.0. Conclusion & Outlook

Mitochondria-inspired metallodrug discovery is a fruitful arena for the development of new medicines. Transition metal complexes possess inherent redox properties that resonate with the redox-active mitochondria organelle. We posit that tuning electrochemical behavior, lipophilicity, and cationic character of metal-complexes are crucial descriptors that influence mitochondria localization and modulation. The geometry, oxidation states, coordination sites, ligand effects, kinetic and thermodynamic characteristics of metal complexes provide an enormous framework for structural diversity that can target distinct mitochondrial targets and pathways. The unique spatial and functional complexes have the potential to unravel new mitochondrial-related molecular targets and mechanisms critical for the treatment of cancer and overcome chemotherapeutic drug resistance, especially platinum drug resistance. To accelerate mitochondrial targeting agents as selective chemical probes or clinically relevant drugs, complexes amenable to optimization and compound libraries for assay screening are needed. One major structural feature depending on the metal core will be to conduct a robust structure activity study that explores the degree of lipophilicity of cationic metal complexes and selective cancer mitochondria accumulation, given the highly negative mitochondria membrane potential of cancer cells. Importantly, relevant biological and disease models are crucial for evaluating compounds. These studies must incorporate selectivity, absorption, distribution, metabolism, excretion, and toxicity parameters in 2D and where possible 3D spheroids or organoids. Employing rigorous mechanism of action studies that factors target identification and validation strategies will be poignant in the development of mitochondria selective anticancer agents. This minireview highlights the potential for selective, metal-based mitochondrial targeting agents using representative examples to ignite excitement and further development.

Acknowledgements

This work was supported by grant R01CA258421-01 from the National Cancer Institute.

Footnotes

Declaration of interests

The authors declare the following financial interests/personal relationships which may be considered as potential competing interests:

Samuel G. Awuah has patent #PCT/US21/52719; PCT/US21/43774; PCT/US21/43766 pending to University of Kentucky Research Foundation.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

Papers of particular interest, published within the period of review, have been highlighted as:

* of special interest

* * of outstanding interest

  • 1.Vyas S, Zaganjor E, Haigis MC: Mitochondria and cancer. Cell 2016, 166:555–566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Wallace DC: Mitochondria and cancer. Nature Reviews Cancer 2012, 12:685–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Warburg O: On the origin of cancer cells. Science 1956, 123:309–314. [DOI] [PubMed] [Google Scholar]
  • 4.Weinhouse S: On Respiratory Impairment in Cancer Cells. Science 1956, 124:267–269. [DOI] [PubMed] [Google Scholar]
  • 5.Weinberg F, Hamanaka R, Wheaton WW, Weinberg S, Joseph J, Lopez M, Kalyanaraman B, Mutlu GM, Budinger GS, Chandel NS: Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proceedings of the National Academy of Sciences 2010, 107:8788–8793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Tan AS, Baty JW, Dong L-F, Bezawork-Geleta A, Endaya B, Goodwin J, Bajzikova M, Kovarova J, Peterka M, Yan B: Mitochondrial genome acquisition restores respiratory function and tumorigenic potential of cancer cells without mitochondrial DNA. Cell metabolism 2015, 21:81–94. [DOI] [PubMed] [Google Scholar]
  • 7.Sullivan LB, Chandel NS: Mitochondrial reactive oxygen species and cancer. Cancer & metabolism 2014, 2:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.DeBerardinis RJ, Mancuso A, Daikhin E, Nissim I, Yudkoff M, Wehrli S, Thompson CB: Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proceedings of the National Academy of Sciences 2007, 104:19345–19350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gasser G, Ott I, Metzler-Nolte N: Organometallic anticancer compounds. Journal of medicinal chemistry 2011, 54:3–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Parveen S, Arjmand F, Tabassum S: Development and future prospects of selective organometallic compounds as anticancer drug candidates exhibiting novel modes of action. European Journal of Medicinal Chemistry 2019, 175:269–286. [DOI] [PubMed] [Google Scholar]
  • 11.Paprocka R, Wiese-Szadkowska M, Janciauskiene S, Kosmalski T, Kulik M, Helmin-Basa A: Latest developments in metal complexes as anticancer agents. Coordination Chemistry Reviews 2022, 452:214307. [Google Scholar]
  • 12.Oun R, Moussa YE, Wheate NJ: The side effects of platinum-based chemotherapy drugs: a review for chemists. Dalton transactions 2018, 47:6645–6653. [DOI] [PubMed] [Google Scholar]
  • 13.Astolfi L, Ghiselli S, Guaran V, Chicca M, Simoni E, Olivetto E, Lelli G, Martini A: Correlation of adverse effects of cisplatin administration in patients affected by solid tumours: A retrospective evaluation. Oncology reports 2013, 29:1285–1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Yeo CI, Ooi KK, Tiekink ER: Gold-based medicine: a paradigm shift in anti-cancer therapy? Molecules 2018, 23:1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Barnard PJ, Berners-Price SJ: Targeting the mitochondrial cell death pathway with gold compounds. Coordination Chemistry Reviews 2007, 251:1889–1902. [Google Scholar]
  • 16.Chakrabortty S, Agrawalla BK, Stumper A, Vegi NM, Fischer S, Reichardt C, Kögler M, Dietzek B, Feuring-Buske M, Buske C: Mitochondria targeted protein-ruthenium photosensitizer for efficient photodynamic applications. Journal of the American Chemical Society 2017, 139:2512–2519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Liu J, Chen Y, Li G, Zhang P, Jin C, Zeng L, Ji L, Chao H: Ruthenium (II) polypyridyl complexes as mitochondria-targeted two-photon photodynamic anticancer agents. Biomaterials 2015, 56:140–153. [DOI] [PubMed] [Google Scholar]
  • 18.Burke CS, Byrne A, Keyes TE: Highly selective mitochondrial targeting by a ruthenium (II) peptide conjugate: Imaging and photoinduced damage of mitochondrial DNA. Angewandte Chemie International Edition 2018, 57:12420–12424. [DOI] [PubMed] [Google Scholar]
  • 19.Skiba J, Bernaś T, Trzybiński D, Woźniak K, Ferraro G, Marasco D, Merlino A, Shafikov MZ, Czerwieniec R, Kowalski K: Mitochondria targeting with luminescent rhenium (I) complexes. Molecules 2017, 22:809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yi S, Lu Z, Zhang J, Wang J, Xie Z, Hou L: Amphiphilic gemini iridium (III) complex as a mitochondria-targeted theranostic agent for tumor imaging and photodynamic therapy. ACS applied materials & interfaces 2019, 11:15276–15289. [DOI] [PubMed] [Google Scholar]
  • 21.Xiong K, Chen Y, Ouyang C, Guan R-L, Ji L-N, Chao H: Cyclometalated iridium (III) complexes as mitochondria-targeted anticancer agents. Biochimie 2016, 125:186–194. [DOI] [PubMed] [Google Scholar]
  • 22.Cao J-J, Zheng Y, Wu X-W, Tan C-P, Chen M-H, Wu N, Ji L-N, Mao Z-W: Anticancer cyclometalated iridium (III) complexes with planar ligands: mitochondrial DNA damage and metabolism disturbance. Journal of Medicinal Chemistry 2019, 62:3311–3322. [DOI] [PubMed] [Google Scholar]
  • 23.Hearn JM, Romero-Canelón I, Munro AF, Fu Y, Pizarro AM, Garnett MJ, McDermott U, Carragher NO, Sadler PJ: Potent organo-osmium compound shifts metabolism in epithelial ovarian cancer cells. Proceedings of the National Academy of Sciences 2015, 112:E3800–E3805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wang F-Y, Tang X-M, Wang X, Huang K-B, Feng H-W, Chen Z-F, Liu Y-N, Liang H: Mitochondria-targeted platinum(II) complexes induce apoptosis-dependent autophagic cell death mediated by ER-stress in A549 cancer cells. European Journal of Medicinal Chemistry 2018, 155:639–650. [DOI] [PubMed] [Google Scholar]
  • 25.Li J, He X, Zou Y, Chen D, Yang L, Rao J, Chen H, Chan MCW, Li L, Guo Z, et al. : Mitochondria-targeted platinum(ii) complexes: dual inhibitory activities on tumor cell proliferation and migration/invasion via intracellular trafficking of β-catenin†. Metallomics 2017, 9:726–733. [DOI] [PubMed] [Google Scholar]
  • 26.Jin S, Guo Y, Song D, Zhu Z, Zhang Z, Sun Y, Yang T, Guo Z, Wang X: Targeting energy metabolism by a platinum (IV) prodrug as an alternative pathway for cancer suppression. Inorganic Chemistry 2019, 58:6507–6516. [DOI] [PubMed] [Google Scholar]
  • 27.Muhammad N, Tan C-P, Muhammad K, Wang J, Sadia N, Pan Z-Y, Ji L-N, Mao Z-W: Mitochondria-targeting monofunctional platinum (II)–lonidamine conjugates for cancer cell de-energization. Inorganic Chemistry Frontiers 2020, 7:4010–4019. [Google Scholar]
  • 28. Zhu Z, Wang Z, Zhang C, Wang Y, Zhang H, Gan Z, Guo Z, Wang X: Mitochondrion-targeted platinum complexes suppressing lung cancer through multiple pathways involving energy metabolism. Chemical science 2019, 10:3089–3095. * The authors modified pyriplatin by incorporating triphenylphosphonium at three different positions of pyriplatin to afford three new complexs that exhibit cytotoxic mechanism of action different from the traditional cisplatin. Importanly, the new complexes are less toxic to normal cells compared to cisplatin.
  • 29.Dhar S, Lippard SJ: Mitaplatin, a potent fusion of cisplatin and the orphan drug dichloroacetate. Proceedings of the National Academy of Sciences 2009, 106:22199–22204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Marrache S, Pathak RK, Dhar S: Detouring of cisplatin to access mitochondrial genome for overcoming resistance. Proceedings of the National Academy of Sciences 2014, 111:10444–10449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Jin S, Hao Y, Zhu Z, Muhammad N, Zhang Z, Wang K, Guo Y, Guo Z, Wang X: Impact of mitochondrion-targeting group on the reactivity and cytostatic pathway of platinum (IV) complexes. Inorganic chemistry 2018, 57:11135–11145. [DOI] [PubMed] [Google Scholar]
  • 32.Su Y, Tu Y, Lin H, Wang M-M, Zhang G-D, Yang J, Liu H-K, Su Z: Mitochondria-targeted Pt (IV) prodrugs conjugated with an aggregation-induced emission luminogen against breast cancer cells by dual modulation of apoptosis and autophagy inhibition. Journal of Inorganic Biochemistry 2022, 226:111653. [DOI] [PubMed] [Google Scholar]
  • 33.Bertrand B, Casini A: A golden future in medicinal inorganic chemistry: the promise of anticancer gold organometallic compounds. Dalton Transactions 2014, 43:4209–4219. [DOI] [PubMed] [Google Scholar]
  • 34.Arojojoye AS, Mertens RT, Ofori S, Parkin SR, Awuah SG: Synthesis, characterization, and antiproliferative activity of novel chiral [QuinoxP* AuCl2]+ complexes. Molecules 2020, 25:5735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ofori S, Gukathasan S, Awuah SG: Gold-Based Pharmacophore Inhibits Intracellular MYC Protein. Chemistry–A European Journal 2021, 27:4168–4175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Kim JH, Ofori S, Parkin S, Vekaria H, Sullivan PG, Awuah SG: Anticancer gold (III)-bisphosphine complex alters the mitochondrial electron transport chain to induce in vivo tumor inhibition. Chemical science 2021, 12:7467–7479. ** In this work, novel anticancer gold (III) complexes were developed. Lead complex AuPhos-89 inhibit energy production in TNBC MDA-MB-231 and extensive inquiry into its mechanism of action revealed modulation of the mitochondrial electron transcport chain. In addition to potent in vivo efficacy, AuPhos 89 inhibits liver metastasis in 4T1 tumor mouse model.
  • 37.Olelewe C, Kim JH, Ofori S, Mertens RT, Gukathasan S, Awuah SG: Gold (III)-P-chirogenic complex induces mitochondrial dysfunction in triple-negative breast cancer. Iscience 2022, 25:104340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Quero J, Ruighi F, Osada J, Gimeno MC, Cerrada E, Rodriguez-Yoldi MJ: Gold (I) Complexes Bearing Alkylated 1, 3, 5-Triaza-7-phosphaadamantane Ligands as Thermoresponsive Anticancer Agents in Human Colon Cells. Biomedicines 2021, 9:1848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hu D, Liu Y, Lai YT, Tong KC, Fung YM, Lok CN, Che CM: Anticancer gold (III) porphyrins target mitochondrial chaperone Hsp60. Angewandte Chemie International Edition 2016, 55:1387–1391. [DOI] [PubMed] [Google Scholar]
  • 40. Babak MV, Chong KR, Rapta P, Zannikou M, Tang HM, Reichert L, Chang MR, Kushnarev V, Heffeter P, Meier-Menches SM: Interfering with Metabolic Profile of Triple-Negative Breast Cancers Using Rationally Designed Metformin Prodrugs. Angewandte Chemie International Edition 2021, 60:13405–13413. ** This work describes the development of new cyclometalated Au(III) complexes bearing metformin or phenformin molecules. Studies with the lead complex (3met) showed that its antiproliferative effect was 6000-fold more than just metformin. This complex disrupts mitochondria metabolism in cancer cells and significantly reduces tumor size in TNBC mouse model.
  • 41.Rackham O, Nichols SJ, Leedman PJ, Berners-Price SJ, Filipovska A: A gold (I) phosphine complex selectively induces apoptosis in breast cancer cells: implications for anticancer therapeutics targeted to mitochondria. biochemical pharmacology 2007, 74:992–1002. [DOI] [PubMed] [Google Scholar]
  • 42.Berners-Price SJ, Girard GR, Hill DT, Sutton BM, Jarrett PS, Faucette LF, Johnson RK, Mirabelli CK, Sadler PJ: Cytotoxicity and antitumor activity of some tetrahedral bis (diphosphino) gold (I) chelates. Journal of medicinal chemistry 1990, 33:1386–1392. [DOI] [PubMed] [Google Scholar]
  • 43. Tong K-C, Lok C-N, Wan P-K, Hu D, Fung YME, Chang X-Y, Huang S, Jiang H, Che C-M: An anticancer gold (III)-activated porphyrin scaffold that covalently modifies protein cysteine thiols. Proceedings of the National Academy of Sciences 2020, 117:1321–1329. ** This work describes the discovery of a new biomolecular interation with metal complexes. Here, it involves gold(III) ion activation of the porphyrin ring towards adduct formation with cysteine thiols found in proteins important for cancer survival
  • 44.Kim JH, Reeder E, Parkin S, Awuah SG: Gold (I/III)-phosphine complexes as potent antiproliferative agents. Scientific reports 2019, 9:1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Fung SK, Zou T, Cao B, Lee PY, Fung YME, Hu D, Lok CN, Che CM: Cyclometalated Gold (III) Complexes Containing N-Heterocyclic Carbene Ligands Engage Multiple Anti-Cancer Molecular Targets. Angewandte Chemie 2017, 129:3950–3954. [DOI] [PubMed] [Google Scholar]
  • 46.Luo H, Cao B, Chan AS, Sun RWY, Zou T: Cyclometalated Gold (III)-hydride complexes exhibit visible light-induced thiol reactivity and act as potent photo-activated anti-cancer agents. Angewandte Chemie 2020, 132:11139–11145. [DOI] [PubMed] [Google Scholar]
  • 47. Hyun Kim J, Ofori S, Mertens RT, Parkin S, Awuah SG: Water-Soluble Gold (III)–Metformin Complex Alters Mitochondrial Bioenergetics in Breast Cancer Cells. ChemMedChem 2021, 16:3222–3230. ** The development of new cyclometalated Au(III) complexes (Auraformin) bearing metformin or phenylmetformin molecule was reported. Auraformin is water soluble and disrupts mitochondria bioenergetics 100-fold better than metformin
  • 48.Elsayed A, Abomhya A, Avdiushko M, Kapur N, Awuah S, Barrett T, Goretsky T: NEW OXPHOS REGULATING COMPOUND–AUPHOS–INCREASES MITOCHONDRIAL COMPLEXES EXPRESSION IN MURINE AND HUMAN INTESTINAL EPITHELIAL CELLS. Inflammatory Bowel Diseases 2022, 28:S51–S51. [Google Scholar]
  • 49.Wempe L, Mohamed R, Warinner J, Goretsky T, Avdiushko M, Kim J, Abomhya A, Lee G, Awuah S, Barrett T: AUPHOS, A “FIRST-IN CLASS” ORAL AGENT FOR CORRECTING METABOLIC DYSFUNCTION IN IBD. Gastroenterology 2022, 162:S2. [Google Scholar]
  • 50.Mohamed R, Warinner J, Wempe L, Avdiushko M, Goretsky T, Kim J, Abomhya A, Lee G, Awuah S, Barrett T: AUPHOS, A NOVEL THERAPEUTIC THAT IMPROVES MITOCHONDRIAL FUNCTION AND AMELIORATES CHRONIC COLITIS. Gastroenterology 2022, 162:S2–S3. [Google Scholar]
  • 51. Mertens RT, Jennings WC, Ofori S, Kim JH, Parkin S, Kwakye GF, Awuah SG: Synthetic Control of Mitochondrial Dynamics: Developing Three-Coordinate Au (I) Probes for Perturbation of Mitochondria Structure and Function. JACS Au 2021, 1:439–449. ** The quest for tool compounds and therapeutics targeting mitochondrial structure has been elusive, despite the important role of mitochondrial organization and structure in physiology and drug resistance. For the first time, the rational development of new tricoordinate gold(I) complexes to modulate mitochondrial structure was reported. Lead complex AuTri-9 perturbs mitochondria structure and is highly tolerable in vivo.
  • 52.Colombo A, Fontani M, Dragonetti C, Roberto D, Williams JG, Scotto di Perrotolo R, Casagrande F, Barozzi S, Polo S: A Highly Luminescent Tetrahydrocurcumin IrIII Complex with Remarkable Photoactivated Anticancer Activity. Chemistry–A European Journal 2019, 25:7948–7952. [DOI] [PubMed] [Google Scholar]
  • 53.Li Y, Tan C-P, Zhang W, He L, Ji L-N, Mao Z-W: Phosphorescent iridium (III)-bis-N-heterocyclic carbene complexes as mitochondria-targeted theranostic and photodynamic anticancer agents. Biomaterials 2015, 39:95–104. [DOI] [PubMed] [Google Scholar]
  • 54.He L, Li Y, Tan C-P, Ye R-R, Chen M-H, Cao J-J, Ji L-N, Mao Z-W: Cyclometalated iridium (III) complexes as lysosome-targeted photodynamic anticancer and real-time tracking agents. Chemical science 2015, 6:5409–5418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Sudheesh KV, Jayaram PS, Samanta A, Bejoymohandas KS, Jayasree RS, Ajayaghosh A: A Cyclometalated IrIII Complex as a Lysosome-Targeted Photodynamic Therapeutic Agent for Integrated Imaging and Therapy in Cancer Cells. Chemistry–A European Journal 2018, 24:10999–11007. [DOI] [PubMed] [Google Scholar]
  • 56.Nam JS, Kang M-G, Kang J, Park S-Y, Lee SJC, Kim H-T, Seo JK, Kwon O-H, Lim MH, Rhee H-W: Endoplasmic reticulum-localized iridium (III) complexes as efficient photodynamic therapy agents via protein modifications. Journal of the American Chemical Society 2016, 138:10968–10977. [DOI] [PubMed] [Google Scholar]
  • 57.Yuan B, Liu J, Guan R, Jin C, Ji L, Chao H: Endoplasmic reticulum targeted cyclometalated iridium (iii) complexes as efficient photodynamic therapy photosensitizers. Dalton Transactions 2019, 48:6408–6415. [DOI] [PubMed] [Google Scholar]
  • 58. Li X, Wu J, Wang L, He C, Chen L, Jiao Y, Duan C: Mitochondrial-DNA-Targeted IrIII-Containing Metallohelices with Tunable Photodynamic Therapy Efficacy in Cancer Cells. Angewandte Chemie International Edition 2020, 59:6420–6427. * This is the first report of new phototoxic dinuclear IrIII-based metallohelices designed to target mitochondria DNA for better photodynamic therapy
  • 59.Kuang S, Wei F, Karges J, Ke L, Xiong K, Liao X, Gasser G, Ji L, Chao H: Photodecaging of a Mitochondria-Localized Iridium (III) Endoperoxide Complex for Two-Photon Photoactivated Therapy under Hypoxia. Journal of the American Chemical Society 2022, 144:4091–4101. [DOI] [PubMed] [Google Scholar]
  • 60.Novohradsky V, Vigueras G, Pracharova J, Cutillas N, Janiak C, Kostrhunova H, Brabec V, Ruiz J, Kasparkova J: Molecular superoxide radical photogeneration in cancer cells by dipyridophenazine iridium (III) complexes. Inorganic Chemistry Frontiers 2019, 6:2500–2513. [Google Scholar]
  • 61.Markova L, Novohradsky V, Kasparkova J, Ruiz J, Brabec V: Dipyridophenazine iridium (III) complex as a phototoxic cancer stem cell selective, mitochondria targeting agent. Chemico-Biological Interactions 2022, 360:109955. [DOI] [PubMed] [Google Scholar]
  • 62.Sancho P, Barneda D, Heeschen C: Hallmarks of cancer stem cell metabolism. British journal of cancer 2016, 114:1305–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. He L, Xiong K, Wang L, Guan R, Chen Y, Ji L, Chao H: Iridium (iii) complexes as mitochondrial topoisomerase inhibitors against cisplatin-resistant cancer cells. Chemical Communications 2021, 57:8308–8311. * Here, the development of the first organometallic mitochondria topoisomerase inhibitors is described.
  • 64.Marker SC, King AP, Granja S, Vaughn B, Woods JJ, Boros E, Wilson JJ: Exploring the in vivo and in vitro anticancer activity of rhenium isonitrile complexes. Inorganic chemistry 2020, 59:10285–10303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Knopf KM, Murphy BL, MacMillan SN, Baskin JM, Barr MP, Boros E, Wilson JJ: In vitro anticancer activity and in vivo biodistribution of rhenium (I) tricarbonyl aqua complexes. Journal of the American Chemical Society 2017, 139:14302–14314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Konkankit CC, King AP, Knopf KM, Southard TL, Wilson JJ: In vivo anticancer activity of a rhenium (I) tricarbonyl complex. ACS medicinal chemistry letters 2019, 10:822–827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Pan ZY, Tan CP, Rao LS, Zhang H, Zheng Y, Hao L, Ji LN, Mao ZW: Recoding the Cancer Epigenome by Intervening in Metabolism and Iron Homeostasis with Mitochondria-Targeted Rhenium (I) Complexes. Angewandte Chemie International Edition 2020, 59:18755–18762. ** This work describes the development and biological effects of a mitochondria targeting rhenium (I) complex bearing deferasirox (a known iron chelating agent). This complex DFX-Re3 relocates intracellular iron into the mitochondria and disrupt mitochondria metabolism. Hence effecting changes to metabolic species involved in epigenetic modifications.
  • 68.Wang F-X, Liang J-H, Zhang H, Wang Z-H, Wan Q, Tan C-P, Ji L-N, Mao Z-W: Mitochondria-accumulating rhenium (I) Tricarbonyl complexes induce cell death via irreversible oxidative stress and glutathione metabolism disturbance. ACS applied materials & interfaces 2019, 11:13123–13133. [DOI] [PubMed] [Google Scholar]
  • 69.Díaz-García JD, Gallegos-Villalobos A, Gonzalez-Espinoza L, Sanchez-Nino MD, Villarrubia J, Ortiz A: Deferasirox nephrotoxicity—the knowns and unknowns. Nature reviews nephrology 2014, 10:574–586. [DOI] [PubMed] [Google Scholar]
  • 70.He S-F, Liao J-X, Huang M-Y, Zhang Y-Q, Zou Y-M, Wu C-L, Lin W-Y, Chen J-X, Sun J: Rhenium–guanidine complex as photosensitizer: trigger HeLa cell apoptosis through death receptor-mediated, mitochondria-mediated, and cell cycle arrest pathways. Metallomics 2022, 14. [DOI] [PubMed] [Google Scholar]
  • 71.Ye R-R, Chen B-C, Lu J-J, Ma X-R, Li R-T: Phosphorescent rhenium(I) complexes conjugated with artesunate: Mitochondrial targeting and apoptosis-ferroptosis dual induction. Journal of Inorganic Biochemistry 2021, 223:111537. [DOI] [PubMed] [Google Scholar]

RESOURCES