Network


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

Hotspot


Dive into the research topics where Peter M. Bruno is active.

Publication


Featured researches published by Peter M. Bruno.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Defining principles of combination drug mechanisms of action.

Justin R. Pritchard; Peter M. Bruno; Luke A. Gilbert; Kelsey L. Capron; Douglas A. Lauffenburger; Michael T. Hemann

Combination chemotherapies have been a mainstay in the treatment of disseminated malignancies for almost 60 y, yet even successful regimens fail to cure many patients. Although their single-drug components are well studied, the mechanisms by which drugs work together in clinical combination regimens are poorly understood. Here, we combine RNAi-based functional signatures with complementary informatics tools to examine drug combinations. This approach seeks to bring to combination therapy what the knowledge of biochemical targets has brought to single-drug therapy and creates a statistical and experimental definition of “combination drug mechanisms of action.” We show that certain synergistic drug combinations may act as a more potent version of a single drug. Conversely, unlike these highly synergistic combinations, most drugs average extant single-drug variations in therapeutic response. When combined to form multidrug regimens, averaging combinations form averaging regimens that homogenize genetic variation in mouse models of cancer and in clinical genomics datasets. We suggest surprisingly simple and predictable combination mechanisms of action that are independent of biochemical mechanism and have implications for biomarker discovery as well as for the development of regimens with defined genetic dependencies.


Journal of the American Chemical Society | 2014

A breast cancer stem cell-selective, mammospheres-potent osmium(VI) nitrido complex.

Kogularamanan Suntharalingam; Wei Lin; Timothy C. Johnstone; Peter M. Bruno; Yao Rong Zheng; Michael T. Hemann; Stephen J. Lippard

The effect of a newly developed osmium(VI) nitrido complex, 1, on breast cancer stem cells (CSCs) is reported. The complex displays selective toxicity for HMLER breast cancer cells enriched with CD44-positive, CSC-like cells over the same cells having reduced CSC character. Remarkably, 1 also reduces the proportion of CSCs within a heterogeneous breast cancer cell population and irreversibly inhibits the formation of free-floating mammospheres to an extent similar to that of salinomycin, a natural product that targets CSCs. Detailed mechanistic studies reveal that in breast cancer cells 1 induces DNA damage and endoplasmic reticulum stress, the latter being responsible for the CSC selectivity. The anti-CSC properties of 1 provide a strong impetus for the development of new metal-based compounds to target CSCs and to treat chemotherapy-resistant and relapsed tumors.


Nature Medicine | 2017

A subset of platinum-containing chemotherapeutic agents kills cells by inducing ribosome biogenesis stress.

Peter M. Bruno; Yunpeng Liu; Ga Young Park; Junko Murai; Catherine E Koch; Timothy J Eisen; Justin R. Pritchard; Yves Pommier; Stephen J. Lippard; Michael T. Hemann

Cisplatin and its platinum analogs, carboplatin and oxaliplatin, are some of the most widely used cancer chemotherapeutics. Although cisplatin and carboplatin are used primarily in germ cell, breast and lung malignancies, oxaliplatin is instead used almost exclusively to treat colorectal and other gastrointestinal cancers. Here we utilize a unique, multi-platform genetic approach to study the mechanism of action of these clinically established platinum anti-cancer agents, as well as more recently developed cisplatin analogs. We show that oxaliplatin, unlike cisplatin and carboplatin, does not kill cells through the DNA-damage response. Rather, oxaliplatin kills cells by inducing ribosome biogenesis stress. This difference in drug mechanism explains the distinct clinical implementation of oxaliplatin relative to cisplatin, and it might enable mechanistically informed selection of distinct platinum drugs for distinct malignancies. These data highlight the functional diversity of core components of front-line cancer therapy and the potential benefits of applying a mechanism-based rationale to the use of our current arsenal of anti-cancer drugs.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Versatile in vivo regulation of tumor phenotypes by dCas9-mediated transcriptional perturbation

Christian Braun; Peter M. Bruno; Max A. Horlbeck; Luke A. Gilbert; Jonathan S. Weissman; Michael T. Hemann

Significance Tumor development is accompanied by widespread genomic and transcriptional changes. The mere acquisition of this information has greatly outpaced our capability to functionally study the biological roles of altered genes. This dilemma highlights the necessity to develop technologies that facilitate a rapid functional prioritization among lists of altered genes. Here, we use catalytically dead Cas9 to specifically activate or inactivate the transcription of genes in mouse models of cancer. This approach allows us to study the impact of gene-level changes in vivo and to systematically screen for novel genetic mediators of treatment relapse. We expect that this approach can be used to systematically dissect the biological role of cancer-related genes, a process critical to identifying new cancer drug targets. Targeted transcriptional regulation is a powerful tool to study genetic mediators of cellular behavior. Here, we show that catalytically dead Cas9 (dCas9) targeted to genomic regions upstream or downstream of the transcription start site allows for specific and sustainable gene-expression level alterations in tumor cells in vitro and in syngeneic immune-competent mouse models. We used this approach for a high-coverage pooled gene-activation screen in vivo and discovered previously unidentified modulators of tumor growth and therapeutic response. Moreover, by using dCas9 linked to an activation domain, we can either enhance or suppress target gene expression simply by changing the genetic location of dCas9 binding relative to the transcription start site. We demonstrate that these directed changes in gene-transcription levels occur with minimal off-target effects. Our findings highlight the use of dCas9-mediated transcriptional regulation as a versatile tool to reproducibly interrogate tumor phenotypes in vivo.


Journal of the American Chemical Society | 2015

Necroptosis-inducing rhenium(V) oxo complexes.

Kogularamanan Suntharalingam; Samuel G. Awuah; Peter M. Bruno; Timothy C. Johnstone; Fang Wang; Wei Lin; Yao Rong Zheng; Julia E. Page; Michael T. Hemann; Stephen J. Lippard

Rhenium(V) oxo complexes of general formula [ReO(OMe)(N^N)Cl2], where N^N = 4,7-diphenyl-1,10-phenanthroline, 1, or 3,4,7,8-tetramethyl-1,10-phenanthroline, 2, effectively kill cancer cells by triggering necroptosis, a non-apoptotic form of cell death. Both complexes evoke necrosome (RIP1-RIP3)-dependent intracellular reactive oxygen species (ROS) production and propidium iodide uptake. The complexes also induce mitochondrial membrane potential depletion, a possible downstream effect of ROS production. Apparently, 1 and 2 are the first rhenium complexes to evoke cellular events consistent with programmed necrosis in cancer cells. Furthermore, 1 and 2 display low acute toxicity in C57BL/6 mice and reasonable stability in fresh human blood.


Angewandte Chemie | 2016

Breast Cancer Stem Cell Potent Copper(II)–Non‐Steroidal Anti‐Inflammatory Drug Complexes

Janine N. Boodram; Iain McGregor; Peter M. Bruno; Paul B. Cressey; Michael T. Hemann; Kogularamanan Suntharalingam

The breast cancer stem cell (CSC) potency of a series of copper(II)-phenanthroline complexes containing the nonsteroidal anti-inflammatory drug (NSAID), indomethacin, is reported. The most effective copper(II) complex in this series, 4, selectivity kills breast CSC-enriched HMLER-shEcad cells over breast CSC-depleted HMLER cells. Furthermore, 4 reduces the formation, size, and viability of mammospheres, to a greater extent than salinomycin, a potassium ionophore known to selectively inhibit CSCs. Mechanistic studies revealed that the CSC-specificity observed for 4 arises from its ability to generate intracellular reactive oxygen species (ROS) and inhibit cyclooxygenase-2 (COX-2), an enzyme that is overexpressed in breast CSCs. The former induces DNA damage, activates JNK and p38 pathways, and leads to apoptosis.


Journal of the American Chemical Society | 2016

Using an RNAi Signature Assay To Guide the Design of Three-Drug-Conjugated Nanoparticles with Validated Mechanisms, In Vivo Efficacy, and Low Toxicity

Jonathan C. Barnes; Peter M. Bruno; Hung V.-T. Nguyen; Longyan Liao; Jenny Liu; Michael T. Hemann; Jeremiah A. Johnson

Single-nanoparticle (NP) combination chemotherapeutics are quickly emerging as attractive alternatives to traditional chemotherapy due to their ability to increase drug solubility, reduce off-target toxicity, enhance blood circulation lifetime, and increase the amount of drug delivered to tumors. In the case of NP-bound drugs, that is, NP-prodrugs, the current standard of practice is to assume that the subcellular mechanism of action for each drug released from the NP mirrors that of the unbound, free-drug. Here, we use an RNAi signature assay for the first time to examine the mechanism of action of multidrug-conjugated NP prodrugs relative to their small molecule prodrugs and native drug mechanisms of action. Additionally, the effective additive contribution of three different drugs in a single-NP platform is characterized. The results indicate that some platinum(IV) cisplatin prodrugs, although cytotoxic, may not have the expected mechanism of action for cisplatin. This insight was utilized to develop a novel platinum(IV) oxaliplatin prodrug and incorporate it into a three-drug-conjugated NP, where each drugs mechanism of action is preserved, to treat tumor-bearing mice with otherwise lethal levels of chemotherapy.


Dalton Transactions | 2016

The breast cancer stem cell potency of copper(II) complexes bearing nonsteroidal anti-inflammatory drugs and their encapsulation using polymeric nanoparticles

Arvin Eskandari; Janine N. Boodram; Paul B. Cressey; Chunxin Lu; Peter M. Bruno; Michael T. Hemann; Kogularamanan Suntharalingam

We report the cancer stem cell (CSC) potency of a novel series of copper(ii)-phenanthroline complexes bearing nonsteriodial anti-inflammatory drugs: naproxen, tolfenamic acid, and indomethacin (2a-3c). Two of the complexes, 2a and 3c, kill breast CSC-enriched HMLER-shEcad cells (grown in both monolayer and three-dimensional cell cultures) to a significantly better extent than salinomycin, a well-established CSC toxin. The most potent complex in the series, 3c induces its cytotoxic effect by generating intracellular reactive oxygen species (ROS) and inhibiting cyclooxgenase-2 (COX-2) activity. Encapsulation of 3c using biodegradable methoxy poly(ethylene glycol)-b-poly(d,l-lactic-co-glycolic) acid (PEG-PLGA) copolymers at the appropriate feed (5%, 3c NP5) enhances breast CSC uptake and reduces overall toxicity. The nanoparticle formulation, 3c NP5 selectively kills breast CSCs over bulk breast cancer cells, and evokes a similar cellular response to the payload, 3c. To the best of our knowledge, this is the first study to demonstrate that polymeric nanoparticles can be used to effectively deliver CSC-potent metal complexes into CSCs.


ChemBioChem | 2016

The Potent Inhibitory Effect of a Naproxen-Appended Cobalt(III)-Cyclam Complex on Cancer Stem Cells

Paul B. Cressey; Arvin Eskandari; Peter M. Bruno; Chunxin Lu; Michael T. Hemann; Kogularamanan Suntharalingam

We report the potency against cancer stem cells (CSCs) of a new cobalt(III)‐cyclam complex (1) that bears the nonsteroidal anti‐inflammatory drug, naproxen. The complex displays selective potency for breast CSC‐enriched HMLER‐shEcad cells over breast CSC‐depleted HMLER cells. Additionally, it inhibited the formation of three‐dimensional tumour‐like mammospheres, and reduced their viability to a greater extent than clinically used breast cancer drugs (vinorelbine, cisplatin and paclitaxel). The anti‐mammosphere potency of 1 was enhanced under hypoxia‐mimicking conditions. Detailed mechanistic studies revealed that DNA damage and cyclooxygenase‐2 (COX‐2) inhibition contribute to the cytotoxic mechanism of 1. To the best of our knowledge, 1 is the first cobalt‐containing compound to show selective potency for CSCs over bulk cancer cells.


Nature Communications | 2017

Nanoparticle conjugates of a highly potent toxin enhance safety and circumvent platinum resistance in ovarian cancer

Ruogu Qi; Yongheng Wang; Peter M. Bruno; Haihua Xiao; Yingjie Yu; Ting Li; Sam Lauffer; Wei Wei; Qixian Chen; Xiang Kang; Haiqin Song; Xi Yang; Xing Huang; Alexandre Detappe; Ursula A. Matulonis; David Pepin; Michael T. Hemann; Michael J. Birrer; P. Peter Ghoroghchian

Advanced-stage epithelial ovarian cancers are amongst the most difficult to treat tumors and have proven to be refractory to most cytotoxic, molecularly targeted, or immunotherapeutic approaches. Here, we report that nanoparticle-drug conjugates (NDCs) of monomethyl auristatin E (MMAE) significantly increase loading on a per-vehicle basis as compared to antibody-drug conjugates (ADCs). Their intraperitoneal administration enabled triggered release of the active MMAE toxin to inhibit tumor growth and to extend animal survival to >90 days in a cell-line xenograft model of disseminated ovarian cancer. In a patient-derived xenograft model of advanced-stage and platinum-resistant ovarian cancer, an MMAE-based NDC doubled the duration of tumor growth inhibition as compared to cisplatin. NDCs of highly potent toxins thus introduce a translatable platform that may be exploited to maximize the safety and efficacy of cytotoxic chemotherapies, combining the best features of ADCs with those of nanoparticle-based therapeutics.Improving the safety and efficacy of chemotherapeutics will help to enhance their effects. Here, the authors show that intraperitoneal delivery of nanoparticle conjugates of a potent toxin prolongs tumor inhibition and survival as compared to cisplatin in advanced-stage and platinum-resistant ovarian cancer mouse models.

Collaboration


Dive into the Peter M. Bruno's collaboration.

Top Co-Authors

Avatar

Michael T. Hemann

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stephen J. Lippard

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Wei Lin

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Timothy C. Johnstone

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Justin R. Pritchard

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Samuel G. Awuah

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge