Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lisa Maria Mustachio is active.

Publication


Featured researches published by Lisa Maria Mustachio.


Cancer Research | 2015

CDK2 Inhibition Causes Anaphase Catastrophe in Lung Cancer through the Centrosomal Protein CP110

Shanhu Hu; Alexey V. Danilov; Kristina M. Godek; Bernardo Orr; Laura J. Tafe; Jaime Rodriguez-Canales; Carmen Behrens; Barbara Mino; Cesar A. Moran; Vincent A. Memoli; Lisa Maria Mustachio; Fabrizio Galimberti; Saranya Ravi; Andrew De Castro; Yun Lu; David Sekula; Angeline S. Andrew; Ignacio I. Wistuba; Sarah J. Freemantle; Duane A. Compton; Ethan Dmitrovsky

Aneuploidy is frequently detected in human cancers and is implicated in carcinogenesis. Pharmacologic targeting of aneuploidy is an attractive therapeutic strategy, as this would preferentially eliminate malignant over normal cells. We previously discovered that CDK2 inhibition causes lung cancer cells with more than two centrosomes to undergo multipolar cell division leading to apoptosis, defined as anaphase catastrophe. Cells with activating KRAS mutations were especially sensitive to CDK2 inhibition. Mechanisms of CDK2-mediated anaphase catastrophe and how activated KRAS enhances this effect were investigated. Live-cell imaging provided direct evidence that following CDK2 inhibition, lung cancer cells develop multipolar anaphase and undergo multipolar cell division with the resulting progeny apoptotic. The siRNA-mediated repression of the CDK2 target and centrosome protein CP110 induced anaphase catastrophe of lung cancer cells. In contrast, CP110 overexpression antagonized CDK2 inhibitor-mediated anaphase catastrophe. Furthermore, activated KRAS mutations sensitized lung cancer cells to CDK2 inhibition by deregulating CP110 expression. Thus, CP110 is a critical mediator of CDK2 inhibition-driven anaphase catastrophe. Independent examination of murine and human paired normal-malignant lung tissues revealed marked upregulation of CP110 in malignant versus normal lung. Human lung cancers with KRAS mutations had significantly lower CP110 expression as compared with KRAS wild-type cancers. Thus, a direct link was found between CP110 and CDK2 inhibitor antineoplastic response. CP110 plays a mechanistic role in response of lung cancer cells to CDK2 inhibition, especially in the presence of activated KRAS mutations.


Oncotarget | 2017

The ISG15-specific protease USP18 regulates stability of PTEN

Lisa Maria Mustachio; Masanori Kawakami; Yun Lu; Jaime Rodriguez-Canales; Barbara Mino; Carmen Behrens; Ignacio I. Wistuba; Neus Bota-Rabassedas; Jun Yu; J. Jack Lee; Jason Roszik; Lin Zheng; Xi Liu; Sarah J. Freemantle; Ethan Dmitrovsky

The ubiquitin-like modifier interferon-stimulated gene 15 (ISG15) is implicated in both oncogenic and tumor suppressive programs. Yet, few ISGylation substrates are known and functionally validated in cancer biology. We previously found specific oncoproteins were substrates of ISGylation and were stabilized by the ISG15-specific deubiquitinase (DUB) ubiquitin specific peptidase 18 (USP18). Using reverse-phase protein arrays (RPPAs), this study reports that engineered loss of the DUB USP18 destabilized the tumor suppressor protein phosphatase and tensin homologue (PTEN) in both murine and human lung cancer cell lines. In contrast, engineered gain of USP18 expression in these same lung cancer cell lines stabilized PTEN protein. Using the protein synthesis inhibitor cycloheximide (CHX), USP18 knockdown was shown to destabilize PTEN whereas USP18 overexpression stabilized PTEN protein. Interestingly, repression of USP18 decreased cytoplasmic PTEN relative to nuclear PTEN protein levels. We sought to identify mechanisms engaged in this PTEN protein destabilization using immunoprecipitation assays and found ISG15 directly conjugated with PTEN. To confirm translational relevance of this work, USP18 and PTEN immunohistochemical expression were compared in comprehensive lung cancer arrays. There was a significant (P < 0.0001) positive correlation and association between PTEN and USP18 protein expression profiles in human lung cancers. Taken together, this study identified PTEN as a previously unrecognized substrate of the ISGylation post-translational modification pathway. The deconjugase USP18 serves as a novel regulator of PTEN stability. This indicates inhibition of ISGylation is therapeutically relevant in cancers.


Molecular Cancer Therapeutics | 2015

Specific CP110 Phosphorylation Sites Mediate Anaphase Catastrophe after CDK2 Inhibition: Evidence for Cooperation with USP33 Knockdown

Shanhu Hu; Yun Lu; Bernardo Orr; Kristina M. Godek; Lisa Maria Mustachio; Masanori Kawakami; David Sekula; Duane A. Compton; Sarah J. Freemantle; Ethan Dmitrovsky

Chromosomal instability (CIN) is a hallmark of solid tumor biology and is implicated in carcinogenesis. Preferentially eliminating malignant cells by targeting CIN and aneuploidy is an attractive antineoplastic strategy. We previously reported that CDK2 antagonism causes lung cancer cells to undergo anaphase catastrophe and apoptosis through inhibition of phosphorylation of the centrosomal protein CP110. Cells with activating KRAS mutations were particularly sensitive to CDK2 inhibition due to downregulation of CP110 protein levels. This study investigated mechanisms of CDK2 antagonism that mediate anaphase catastrophe via changes in CP110 protein expression and how activated KRAS affects CP110 levels in lung cancers. Site-directed mutagenesis revealed candidate CDK phosphorylation sites of CP110 (residues Ser 170 and Thr 194) critical for conferring anaphase catastrophe by altering centrosome clustering in mitosis. Intriguingly, KRAS mutation can promote CP110 protein degradation by upregulating the ubiquitin ligase SCFcyclinF, which targets CP110 protein for destabilization. Finally, CDK2 inhibitor response was enhanced when combined with knockdown of the deubiquitinase USP33 that in turn accelerates CP110 protein degradation. Thus, this study provides molecular pharmacologic insights into how CP110 expression regulates response to CDK2 inhibition. An improved understanding of in vitro antineoplastic mechanisms of combining CDK2 antagonism with induced CP110 repression provides a rationale for exploring clinical consequences of this strategy. Taken together, preclinical findings obtained from combining CDK2 inhibition with USP33 repression have implications for treating patients with non–small cell lung cancers. Mol Cancer Ther; 14(11); 2576–85. ©2015 AACR.


Molecular Cancer Therapeutics | 2016

Dinaciclib induces anaphase catastrophe in lung cancer cells via inhibition of cyclin-dependent kinases 1 and 2

Alexey V. Danilov; Shanhu Hu; Bernardo Orr; Kristina M. Godek; Lisa Maria Mustachio; David Sekula; Xi Liu; Masanori Kawakami; Faye M. Johnson; Duane A. Compton; Sarah J. Freemantle; Ethan Dmitrovsky

Despite advances in targeted therapy, lung cancer remains the most common cause of cancer-related mortality in the United States. Chromosomal instability is a prominent feature in lung cancer and, because it rarely occurs in normal cells, it represents a potential therapeutic target. Our prior work discovered that lung cancer cells undergo anaphase catastrophe in response to inhibition of cyclin-dependent kinase 2 (CDK2), followed by apoptosis and reduced growth. In this study, the effects and mechanisms of the multi-CDK inhibitor dinaciclib on lung cancer cells were investigated. We sought to determine the specificity of CDK-dependent induction of anaphase catastrophe. Live cell imaging provided direct evidence that dinaciclib caused multipolar cell divisions resulting in extensive chromosome missegregation. Genetic knockdown of dinaciclib CDK targets revealed that repression of CDK2 and CDK1, but not CDK5 or CDK9, triggered anaphase catastrophe in lung cancer cells. Overexpression of CP110, which is a mediator of CDK2 inhibitor–induced anaphase catastrophe (and a CDK1 and 2 phosphorylation substrate), antagonized anaphase catastrophe and apoptosis following dinaciclib treatment. Consistent with our previous findings, acquisition of activated KRAS sensitized lung cancer cells to dinaciclib-mediated anaphase catastrophe and cell death. Combining dinaciclib with the mitotic inhibitor taxol augmented anaphase catastrophe induction and reduced cell viability of lung cancer cells. Thus, the multi-CDK inhibitor dinaciclib causes anaphase catastrophe in lung cancer cells and should be investigated as a potential therapeutic for wild-type and KRAS-mutant lung cancer, individually or in combination with taxanes. Mol Cancer Ther; 15(11); 2758–66. ©2016 AACR.


Journal of the National Cancer Institute | 2017

Next-Generation CDK2/9 Inhibitors and Anaphase Catastrophe in Lung Cancer

Masanori Kawakami; Lisa Maria Mustachio; Jaime Rodriguez-Canales; Barbara Mino; Jason Roszik; Pan Tong; Jing Wang; J. Jack Lee; Ja Hye Myung; John V. Heymach; Faye M. Johnson; Seungpyo Hong; Lin Zheng; Shanhu Hu; Pamela Villalobos; Carmen Behrens; Ignacio I. Wistuba; Sarah J. Freemantle; Xi Liu; Ethan Dmitrovsky

Background The first generation CDK2/7/9 inhibitor seliciclib (CYC202) causes multipolar anaphase and apoptosis in lung cancer cells with supernumerary centrosomes (known as anaphase catastrophe). We investigated a new and potent CDK2/9 inhibitor, CCT68127 (Cyclacel). Methods CCT68127 was studied in lung cancer cells (three murine and five human) and control murine pulmonary epithelial and human immortalized bronchial epithelial cells. Robotic CCT68127 cell-based proliferation screens were used. Cells undergoing multipolar anaphase and inhibited centrosome clustering were scored. Reverse phase protein arrays (RPPAs) assessed CCT68127 effects on signaling pathways. The function of PEA15, a growth regulator highlighted by RPPAs, was analyzed. Syngeneic murine lung cancer xenografts (n = 4/group) determined CCT68127 effects on tumorigenicity and circulating tumor cell levels. All statistical tests were two-sided. Results CCT68127 inhibited growth up to 88.5% (SD = 6.4%, P < .003) at 1 μM, induced apoptosis up to 42.6% (SD = 5.5%, P < .001) at 2 μM, and caused G1 or G2/M arrest in lung cancer cells with minimal effects on control cells (growth inhibition at 1 μM: 10.6%, SD = 3.6%, P = .32; apoptosis at 2 μM: 8.2%, SD = 1.0%, P = .22). A robotic screen found that lung cancer cells with KRAS mutation were particularly sensitive to CCT68127 ( P = .02 for IC 50 ). CCT68127 inhibited supernumerary centrosome clustering and caused anaphase catastrophe by 14.1% (SD = 3.6%, P < .009 at 1 μM). CCT68127 reduced PEA15 phosphorylation by 70% (SD = 3.0%, P = .003). The gain of PEA15 expression antagonized and its loss enhanced CCT68127-mediated growth inhibition. CCT68127 reduced lung cancer growth in vivo ( P < .001) and circulating tumor cells ( P = .004). Findings were confirmed with another CDK2/9 inhibitor, CYC065. Conclusions Next-generation CDK2/9 inhibition elicits marked antineoplastic effects in lung cancer via anaphase catastrophe and reduced PEA15 phosphorylation.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Polo-like kinase 4 inhibition produces polyploidy and apoptotic death of lung cancers

Masanori Kawakami; Lisa Maria Mustachio; Lin Zheng; Yulong Chen; Jaime Rodriguez-Canales; Barbara Mino; Jonathan M. Kurie; Jason Roszik; Pamela Villalobos; Kelsie L. Thu; Jennifer Silvester; David W. Cescon; Ignacio I. Wistuba; Tak W. Mak; Xi Liu; Ethan Dmitrovsky

Significance Despite current treatments, lung cancers remain a major public health problem. Innovative ways are needed to treat or prevent these cancers. Centrosomes are critical for fidelity of mitosis. Abnormal centrosome numbers can cause aberrant mitosis and cell death. Polo-like kinase 4 (PLK4) regulates centriole duplication, and its deregulation alters centrosome number and mitosis. The potent PLK4 inhibitor CFI-400945 is reported here to exert marked antineoplastic effects against lung cancers. CDK2 inhibition also deregulates mitosis and was found to cooperate with PLK4 antagonism. CFI-400945 is now undergoing phase I clinical trial testing (NCT01954316). Taken together, targeting PLK4 for inhibition holds promise in lung cancer therapy either as a single agent or when combined with an agent that deregulates mitosis. Polo-like kinase 4 (PLK4) is a serine/threonine kinase regulating centriole duplication. CFI-400945 is a highly selective PLK4 inhibitor that deregulates centriole duplication, causing mitotic defects and death of aneuploid cancers. Prior work was substantially extended by showing CFI-400945 causes polyploidy, growth inhibition, and apoptotic death of murine and human lung cancer cells, despite expression of mutated KRAS or p53. Analysis of DNA content by propidium iodide (PI) staining revealed cells with >4N DNA content (polyploidy) markedly increased after CFI-400945 treatment. Centrosome numbers and mitotic spindles were scored. CFI-400945 treatment produced supernumerary centrosomes and mitotic defects in lung cancer cells. In vivo antineoplastic activity of CFI-400945 was established in mice with syngeneic lung cancer xenografts. Lung tumor growth was significantly inhibited at well-tolerated dosages. Phosphohistone H3 staining of resected lung cancers following CFI-400945 treatment confirmed the presence of aberrant mitosis. PLK4 expression profiles in human lung cancers were explored using The Cancer Genome Atlas (TCGA) and RNA in situ hybridization (RNA ISH) of microarrays containing normal and malignant lung tissues. PLK4 expression was significantly higher in the malignant versus normal lung and conferred an unfavorable survival (P < 0.05). Intriguingly, cyclin dependent kinase 2 (CDK2) antagonism cooperated with PLK4 inhibition. Taken together, PLK4 inhibition alone or as part of a combination regimen is a promising way to combat lung cancer.


Cancer Research | 2018

Evidence for the ISG15-specific deubiquitinase usp18 as an antineoplastic target

Lisa Maria Mustachio; Yun Lu; Masanori Kawakami; Jason Roszik; Sarah J. Freemantle; Xi Liu; Ethan Dmitrovsky

Ubiquitination and ubiquitin-like posttranslational modifications (PTM) regulate activity and stability of oncoproteins and tumor suppressors. This implicates PTMs as antineoplastic targets. One way to alter PTMs is to inhibit activity of deubiquitinases (DUB) that remove ubiquitin or ubiquitin-like proteins from substrate proteins. Roles of DUBs in carcinogenesis have been intensively studied, yet few inhibitors exist. Prior work provides a basis for the ubiquitin-specific protease 18 (USP18) as an antineoplastic target. USP18 is the major DUB that removes IFN-stimulated gene 15 (ISG15) from conjugated proteins. Prior work discovered that engineered loss of USP18 increases ISGylation and in contrast to its gain decreases cancer growth by destabilizing growth-regulatory proteins. Loss of USP18 reduced cancer cell growth by triggering apoptosis. Genetic loss of USP18 repressed cancer formation in engineered murine lung cancer models. The translational relevance of USP18 was confirmed by finding its expression was deregulated in malignant versus normal tissues. Notably, the recent elucidation of the USP18 crystal structure offers a framework for developing an inhibitor to this DUB. This review summarizes strong evidence for USP18 as a previously unrecognized pharmacologic target in oncology. Cancer Res; 78(3); 587-92. ©2018 AACR.


Molecular Cancer Research | 2017

Deubiquitinase USP18 Loss Mislocalizes and Destabilizes KRAS in Lung Cancer

Lisa Maria Mustachio; Yun Lu; Laura J. Tafe; Vincent A. Memoli; Jaime Rodriguez-Canales; Barbara Mino; Pamela Villalobos; Ignacio I. Wistuba; Hiroyuki Katayama; Samir M. Hanash; Jason Roszik; Masanori Kawakami; Kwang Jin Cho; John F. Hancock; Fadzai Chinyengetere; Shanhu Hu; Xi Liu; Sarah J. Freemantle; Ethan Dmitrovsky

KRAS is frequently mutated in lung cancers and is associated with aggressive biology and chemotherapy resistance. Therefore, innovative approaches are needed to treat these lung cancers. Prior work implicated the IFN-stimulated gene 15 (ISG15) deubiquitinase (DUB) USP18 as having antineoplastic activity by regulating lung cancer growth and oncoprotein stability. This study demonstrates that USP18 affects the stability of the KRAS oncoprotein. Interestingly, loss of USP18 reduced KRAS expression, and engineered gain of USP18 expression increased KRAS protein levels in lung cancer cells. Using the protein synthesis inhibitor cycloheximide, USP18 knockdown significantly reduced the half-life of KRAS, but gain of USP18 expression significantly increased its stability. Intriguingly, loss of USP18 altered KRAS subcellular localization by mislocalizing KRAS from the plasma membrane. To explore the biologic consequences, immunohistochemical (IHC) expression profiles of USP18 were compared in lung cancers of KrasLA2/+ versus cyclin E engineered mouse models. USP18 expression was higher in Kras-driven murine lung cancers, indicating a link between KRAS and USP18 expression in vivo. To solidify this association, loss of Usp18 in KrasLA2/+/Usp18−/− mice was found to significantly reduce lung cancers as compared with parental KrasLA2/+ mice. Finally, translational relevance was confirmed in a human lung cancer panel by showing that USP18 IHC expression was significantly higher in KRAS-mutant versus wild-type lung adenocarcinomas. Implications: Taken together, this study highlights a new way to combat the oncogenic consequences of activated KRAS in lung cancer by inhibiting the DUB USP18. Mol Cancer Res; 15(7); 905–14. ©2017 AACR.


Molecular Cancer Therapeutics | 2018

Engaging Anaphase Catastrophe Mechanisms to Eradicate Aneuploid Cancers

Masanori Kawakami; Lisa Maria Mustachio; Xi Liu; Ethan Dmitrovsky

Cancer cells often have supernumerary centrosomes that promote genomic instability, a pathognomonic feature of cancer. During mitosis, cancer cells with supernumerary centrosomes undergo bipolar cell division by clustering centrosomes into two poles. When supernumerary centrosome clustering is antagonized, cancer cells are forced to undergo multipolar division leading to death of daughter cells. This proapoptotic pathway, called anaphase catastrophe, preferentially eliminates aneuploid cancer cells and malignant tumors in engineered mouse models. Anaphase catastrophe occurs through the loss or inhibition of the centrosomal protein CP110, a direct cyclin-dependent kinase 1 (CDK1) and CDK2 target. Intriguingly, CP110 is repressed by the KRAS oncoprotein. This sensitizes KRAS-driven lung cancers (an unmet medical need) to respond to CDK2 inhibitors. Anaphase catastrophe-inducing agents like CDK1 and CDK2 antagonists are lethal to cancer cells with supernumerary centrosomes, but can relatively spare normal cells with two centrosomes. This mechanism is proposed to provide a therapeutic window in the cancer clinic following treatment with a CDK1 or CDK2 inhibitor. Taken together, anaphase catastrophe is a clinically tractable mechanism that promotes death of neoplastic tumors with aneuploidy, a hallmark of cancer. Mol Cancer Ther; 17(4); 724–31. ©2018 AACR.


Cancer Research | 2017

Abstract 128: The next-generation CDK2/9 inhibitor CYC065 elicits marked antineoplastic effects in lung cancer by engaging antimetastatic pathways

Masanori Kawakami; Jason Roszik; Lin Zheng; Jonathan M. Kurie; Lisa Maria Mustachio; Xi Liu; Ethan Dmitrovsky

We previously reported CDK2 antagonism with the first generation CDK2/9/7 inhibitor seliciclib (CYC202; Cyclacel) triggered anaphase catastrophe. This occurred when genomically unstable cancer cells with supernumerary centrosomes (a hallmark of cancer) fail to cluster excessive centrosomes at mitosis. This causes multipolar cell division and apoptotic death. Anaphase catastrophe is conferred after CDK2 antagonism of aneuploid cancer cells, sparing bipolar normal cells with two centrosomes. CYC065 (Cyclacel) is a next-generation CDK2/9 inhibitor that is undergoing clinical trial. Here, we explored CYC065 activity against lung cancer cells, some with known high metastatic potential. CYC065 substantially inhibited growth, triggered apoptosis, and induced anaphase catastrophe in murine (ED1, LKR13, and 393P) and human (Hop62, A549, and H1299) lung cancer cells. In marked contrast, these effects were largely unseen in bipolar immortalized pulmonary epithelial (murine C10 and human BEAS-2B) cells. We sought to explore whether CYC065 antineoplastic effects engaged antimetastatic pathways. In vitro migration and invasion assays were performed. CYC065 markedly inhibited migration and invasion of lung cancer cells (murine: 344SQ and KC2; human: A549 and H1299). Reverse Phase Protein Arrays (RPPAs) interrogated nearly 300 growth-regulatory proteins in murine (344SQ and KC2) and human (A549 and H1299) lung cancer cells over time (6, 12, 24, and 48 hours) after CYC065 or vehicle treatments. These lung cancer cells have high metastatic as well migration or invasive potentials. When highlighted proteins were clustered after CYC065 treatment, some species were clustered as significantly up-regulated or down-regulated in all cells over studied time points. Not surprisingly, up-regulated proteins included those involved in DNA damage or apoptosis. In addition to known CDK targets like phosphorylated retinoblastoma protein, novel proteins were markedly down-regulated, including mTOR pathway and integrin pathway proteins such as FAK phosphorylation and Src phosphorylation. Affected pathways were identified using Ingenuity Pathway Analysis (IPA). IPA revealed up-regulation of pathways that engaged ATM signaling, G2/M DNA damage checkpoint regulation, or apoptosis signaling. Down-regulated pathways affected mTOR signaling, cyclins, cell cycle regulation, or integrin pathways. Mouse studies will be presented that examine in vivo effects of CYC065 in reducing metastases. Taken together, the next-generation CDK2/9 inhibitor, CYC065, elicits marked antineoplastic effects by antagonizing migration and invasion of lung cancer cells. Comprehensive RPPA and IPA studies found that distinct pathways trigger these effects. Citation Format: Masanori Kawakami, Jason Roszik, Lin Zheng, Jonathan Kurie, Lisa Maria Mustachio, Xi Liu, Ethan Dmitrovsky. The next-generation CDK2/9 inhibitor CYC065 elicits marked antineoplastic effects in lung cancer by engaging antimetastatic pathways [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 128. doi:10.1158/1538-7445.AM2017-128

Collaboration


Dive into the Lisa Maria Mustachio's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masanori Kawakami

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xi Liu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jason Roszik

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ignacio I. Wistuba

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jaime Rodriguez-Canales

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge