Network


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

Hotspot


Dive into the research topics where Michele Mayo is active.

Publication


Featured researches published by Michele Mayo.


Bioconjugate Chemistry | 2010

Tumor Delivery and In Vivo Processing of Disulfide-Linked and Thioether-Linked Antibody−Maytansinoid Conjugates

Hans K. Erickson; Wayne C. Widdison; Michele Mayo; Kathleen R. Whiteman; Charlene Audette; Sharon D. Wilhelm; Rajeeva Singh

Antibody-drug conjugates (ADCs) are designed to eradicate cancer cells that express the target antigen on their cell surface. A key component of an ADC is the linker that covalently connects the cytotoxic agent to the antibody. Several antibody-maytansinoid conjugates prepared with disulfide-based linkers such as those targeting the CanAg antigen have been shown to display more activity in preclinical mouse xenograft models than corresponding conjugates prepared with uncleavable thioether-based linkers. To investigate how the linker influences delivery and activation of antibody-maytansinoid conjugates, we isolated and characterized the [(3)H]maytansinoids from CanAg-positive tumor tissues following a single intravenous administration of 300 microg/kg (based on maytansinoid dose) of anti-CanAg antibody (huC242)-(3)H-maytansinoid conjugates prepared with cleavable disulfide linkers and an uncleavable thioether linker. We identified three target-dependent tumor metabolites of the disulfide-linked huC242-SPDB-DM4, namely, lysine-N(epsilon)-SPDB-DM4, DM4, and S-methyl-DM4. We found similar metabolites for the less hindered disulfide-linked huC242-SPP-DM1 conjugate with the exception that no S-methyl-DM1 was detected. The sole metabolite of the uncleavable thioether-linked huC242-SMCC-DM1 was lysine-N(epsilon)-SMCC-DM1. The AUC for the metabolites of huC242-SMCC-DM1 at the tumor over 7 d was about 2-fold greater than the corresponding AUC for the metabolites of the disulfide-linked conjugates. The lipophilic metabolites of the disulfide-linked conjugates were found to be nearly 1000 times more cytotoxic than the more hydrophilic lysine-N(epsilon)-linker-maytansinoids in cell-based viability assays when added extracellularly. The cell killing properties associated with the lipophilic metabolites of the disulfide-linked conjugates (DM4 and S-methyl-DM4, and DM1) provide an explanation for the superior in vivo efficacy that is often observed with antibody-maytansinoid conjugates prepared with disulfide-based linkers in xenograft mouse models.


Cancer Research | 2010

Antibody-Maytansinoid Conjugates Designed to Bypass Multidrug Resistance

Yelena Kovtun; Charlene Audette; Michele Mayo; Gregory Jones; Heather Doherty; Erin Maloney; Hans K. Erickson; Xiuxia Sun; Sharon D. Wilhelm; Olga Ab; Katharine C. Lai; Wayne C. Widdison; Brenda Kellogg; Holly Johnson; Jan Pinkas; Robert J. Lutz; Rajeeva Singh; Victor S. Goldmacher; Ravi V. J. Chari

Conjugation of cytotoxic compounds to antibodies that bind to cancer-specific antigens makes these drugs selective in killing cancer cells. However, many of the compounds used in such antibody-drug conjugates (ADC) are substrates for the multidrug transporter MDR1. To evade the MDR1-mediated resistance, we conjugated the highly cytotoxic maytansinoid DM1 to antibodies via the maleimidyl-based hydrophilic linker PEG(4)Mal. Following uptake into target cells, conjugates made with the PEG(4)Mal linker were processed to a cytotoxic metabolite that was retained by MDR1-expressing cells better than a metabolite of similar conjugates prepared with the nonpolar linker N-succinimidyl-4-(maleimidomethyl)cyclohexane-1-carboxylate (SMCC). In accord, PEG(4)Mal-linked conjugates were more potent in killing MDR1-expressing cells in culture. In addition, PEG(4)Mal-linked conjugates were markedly more effective in eradicating MDR1-expressing human xenograft tumors than SMCC-linked conjugates while being tolerated similarly, thus showing an improved therapeutic index. This study points the way to the development of ADCs that bypass multidrug resistance.


Bioconjugate Chemistry | 2011

Design of Antibody−Maytansinoid Conjugates Allows for Efficient Detoxification via Liver Metabolism

Xiuxia Sun; Wayne C. Widdison; Michele Mayo; Sharon D. Wilhelm; Barbara A. Leece; Ravi V. J. Chari; Rajeeva Singh; Hans K. Erickson

Antibody-maytansinoid conjugates (AMCs) are targeted chemotherapeutic agents consisting of a potent microtubule-depolymerizing maytansinoid (DM1 or DM4) attached to lysine residues of a monoclonal antibody (mAb) using an uncleavable thioether linker or a stable disulfide linker. Most of the administered dose of an antibody-based therapeutic is slowly catabolized by the liver and other tissues of the reticuloendothelial system. Maytansinoids released from an AMC during this catabolic process could potentially be a source of toxicity. To investigate this, we isolated and identified liver metabolites in mice for three different [(3)H]AMCs with structures similar to those currently undergoing evaluation in the clinic. We then synthesized each metabolite to confirm the identification and assessed their cytotoxic potencies when added extracellularly. We found that the uncleavable mAb-SMCC-[(3)H]DM1 conjugate was degraded to a single major maytansinoid metabolite, lysine-SMCC-[(3)H]DM1, that was nearly 50-fold less cytotoxic than maytansine. The two disulfide-linked conjugates, mAb-SPP-[(3)H]DM1 and mAb-SPDB-[(3)H]DM4, were also found to be catabolized to the analogous lysine-linked maytansinoid metabolites. However, subsequent reduction, S-methylation, and NADPH-dependent oxidation steps in the liver yielded the corresponding S-methyl sulfoxide and S-methyl sulfone derivatives. The cytotoxic potencies of the oxidized maytansinoids toward several human carcinoma cell lines were found to be 5- to 50-fold less potent than maytansine. Our results suggest that liver plays an important role in the detoxification of both cleavable and uncleavable AMCs.


Blood | 2013

A novel anti-CD37 antibody-drug conjugate with multiple anti-tumor mechanisms for the treatment of B-cell malignancies

Jutta Deckert; Peter U. Park; Sharon Chicklas; Yong Yi; Min Li; Katharine C. Lai; Michele Mayo; Christina N. Carrigan; Hans K. Erickson; Jan Pinkas; Robert J. Lutz; Thomas Chittenden; John M. Lambert

CD37 has gathered renewed interest as a therapeutic target in non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia (CLL); however, CD37-directed antibody-drug conjugates (ADCs) have not been explored. Here, we identified a novel anti-CD37 antibody, K7153A, with potent in vitro activity against B-cell lines through multiple mechanisms including apoptosis induction, antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and complement-dependent cytotoxicity. The antibody was conjugated to the maytansinoid, DM1, a potent antimicrotubule agent, via the thioether linker, N-succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC), and the resulting ADC, IMGN529, retained the intrinsic antibody activities and showed enhanced cytotoxic activity from targeted payload delivery. In lymphoma cell lines, IMGN529 induced G2/M cell cycle arrest after internalization and lysosomal processing to lysine-N(ε)-SMCC-DM1 as the sole intracellular maytansinoid metabolite. IMGN529 was highly active against subcutaneous B-cell tumor xenografts in severe combined immunodeficient mice with comparable or better activity than rituximab, a combination of cyclophosphamide, vincristine, and prednisone, or bendamustine. In human blood cells, CD37 is expressed in B cells at similar levels as CD20, and IMGN529 resulted in potent and specific depletion of normal and CLL B cells. These results support evaluation of the CD37-targeted ADC, IMGN529, in clinical trials in patients with B-cell malignancies including NHL and CLL.


Molecular Cancer Therapeutics | 2016

A New Class of Antibody-Drug Conjugates with Potent DNA Alkylating Activity.

Michael L. Miller; Nathan Fishkin; Wei Li; Kathleen R. Whiteman; Yelena Kovtun; Emily E. Reid; Katie Archer; Erin Maloney; Charlene Audette; Michele Mayo; Alan Wilhelm; Holly A. Modafferi; Rajeeva Singh; Jan Pinkas; Victor S. Goldmacher; John M. Lambert; Ravi V. J. Chari

The promise of tumor-selective delivery of cytotoxic agents in the form of antibody–drug conjugates (ADC) has now been realized, evidenced by the approval of two ADCs, both of which incorporate highly cytotoxic tubulin-interacting agents, for cancer therapy. An ongoing challenge remains in identifying potent agents with alternative mechanisms of cell killing that can provide ADCs with high therapeutic indices and favorable tolerability. Here, we describe the development of a new class of potent DNA alkylating agents that meets these objectives. Through chemical design, we changed the mechanism of action of our novel DNA cross-linking agent to a monofunctional DNA alkylator. This modification, coupled with linker optimization, generated ADCs that were well tolerated in mice and demonstrated robust antitumor activity in multiple tumor models at doses 1.5% to 3.5% of maximally tolerated levels. These properties underscore the considerable potential of these purpose-created, unique DNA-interacting conjugates for broadening the clinical application of ADC technology. Mol Cancer Ther; 15(8); 1870–8. ©2016 AACR.


mAbs | 2014

Lorvotuzumab mertansine, a CD56-targeting antibody-drug conjugate with potent antitumor activity against small cell lung cancer in human xenograft models

Kathleen R. Whiteman; Holly Johnson; Michele Mayo; Charlene Audette; Christina N. Carrigan; Alyssa LaBelle; Lawrence R. Zukerberg; John M. Lambert; Robert J. Lutz

Lorvotuzumab mertansine (LM) is an antibody-drug conjugate composed of a humanized anti-CD56 antibody, lorvotuzumab, linked via a cleavable disulfide linker to the tubulin-binding maytansinoid DM1. CD56 is expressed on most small cell lung cancers (SCLC), providing a promising therapeutic target for treatment of this aggressive cancer, which has a poor five-year survival rate of only 5–10%. We performed immunohistochemical staining on SCLC tumor microarrays, which confirmed that CD56 is expressed at high levels on most (~74%) SCLC tumors. Conjugation of lorvotuzumab with DM1 did not alter its specific binding to cells and LM demonstrated potent target-dependent cytotoxicity against CD56-positive SCLC cells in vitro. The anti-tumor activity of LM was evaluated against SCLC xenograft models in mice, both as monotherapy and in combination with platinum/etoposide and paclitaxel/carboplatin. Dose-dependent and antigen-specific anti-tumor activity of LM monotherapy was demonstrated at doses as low as 3 mg/kg. LM was highly active in combination with standard-of-care platinum/etoposide therapies, even in relatively resistant xenograft models. LM demonstrated outstanding anti-tumor activity in combination with carboplatin/etoposide, with superior activity over chemotherapy alone when LM was used in combinations at significantly reduced doses (6-fold below the minimally efficacious dose for LM monotherapy). The combination of LM with carboplatin/paclitaxel was also highly active. This study provides the rationale for clinical evaluation of LM as a promising novel targeted therapy for SCLC, both as monotherapy and in combination with chemotherapy.


Molecular Cancer Therapeutics | 2009

Abstract B126: Potent antigen‐specific anti‐tumor activity observed with antibody‐drug conjugates (ADCs) made using a new class of DNA‐crosslinking agents

Michael L. Miller; Nathan Fishkin; Wei Li; Barbara A. Leece; Michele Mayo; Gregory Jones; Emily E. Reid; Katie Archer; Erin Maloney; Yelena Kovtun; Jan Pinkas; Rajeeva Singh; Ravi V. J. Chari

The clinical support for ADC therapeutics has expanded as more highly‐engineered ADCs advance in human clinical testing. Most of the ADCs now in clinical testing contain a tubulin‐acting compound (a maytansine or dolastatin derivative) as the cytotoxic agent. While tubulin‐acting agents can be effective against many different types of cancers, some cancers are more responsive to DNA‐acting agents. To expand the therapeutic potential for ADCs, we sought to develop a new class of cytotoxic agents with a novel, DNA‐acting mechanism of action for use with tumor‐targeting antibodies. Herein, we report the development of our IGN family of cytotoxic agents. These IGN agents comprise indolino‐benzodiazepine dimers that are highly potent by virtue of their ability to alkylate and crosslink DNA. This novel class of compounds demonstrated sequence‐selective DNA adduct formation in vitro and cytotoxicity in the picomolar range towards cultured human cancer cells. The intense potency of these compounds, along with their desired aqueous solubility and stability, make them ideally suited for use in ADCs. A lead compound from this class was conjugated to an EpCAM‐binding antibody, B38.1, and to a CD33‐binding antibody, huMy9‐6, through amide bonds. The B38.1‐IGN conjugate was highly potent against three different EpCAM‐expressing cell lines ‐ COLO 205, LoVo and OVCAR‐3 ‐ with IC50 values of 1 pM, 5 pM and 18 pM, respectively. The addition of excess unconjugated B38.1 antibody abolished this cytotoxic effect, demonstrating that the activity of the conjugate is antigen specific. The B38.1‐IGN conjugate was considerably less potent towards the antigen‐negative Namalwa cell line, with an IC50 value of >1 nM, further demonstrating antigen specificity. Similar potent cytotoxicity was seen with a huMy9‐6‐IGN conjugate targeting the CD33‐positive human promyelocytic leukemia cell line, NB4 (IC50 ∼4pM), in spite of the low antigen expression level (∼10,000 molecules/cell) in this cell line. Of particular interest, the B38.1‐IGN conjugate also was potent towards multidrug resistant cancer cells. B38.1‐IGN had a IC50 value of 14 pM for COLO 205MDR, a COLO 205 clone engineered to overexpress MDR1 transporter, and 7 pM for HCT‐15, an EpCAM‐expressing cell line that naturally expresses MDR1. Antibody‐IGN conjugates demonstrated dose‐dependent activity in multiple human tumor xenograft models in mice, with anti‐tumor activity observed at non‐toxic doses. The unique mechanism of action of the IGN class of compounds, and the high antigen‐specific potency of antibody‐IGN conjugates seen in vitro and in vivo, provides a promising new cytotoxic agent for use in the development of new ADCs. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):B126.


Cancer Research | 2011

Abstract 2830: Antibody and linker selection for the anti-CD37 antibody-maytansinoid conjugate IMGN529 for the treatment of B-cell malignancies

Peter U. Park; Yong Yi; Min Li; Sharon Chicklas; Katharine C. Lai; Michele Mayo; Jennifer Coccia; Jutta Deckert; Jan Pinkas; Thomas D. Chittenden

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL CD37 represents an attractive target for an antibody-maytansinoid conjugate (AMC) due to its prevalence in B-cell malignancies, such as non-Hodgkins lymphoma (NHL) and chronic lymphocytic leukemia (CLL), and its restricted expression on normal tissue, where it is mainly found on B-cells in blood and lymphoid tissues. Additionally, since antibodies to CD37 have been described to have anti-tumor activity, this target has potential for the development of an AMC containing a functional antibody. To select the antibody for this AMC, a large panel of anti-CD37 antibodies was generated by immunizing mice with CD37+ cells. Anti-CD37 antibodies were selected based on their superior ability to induce apoptosis in Ramos and Raji cells in comparison to the anti-CD20 antibody, rituximab, and the anti-CD37 SMIP, TRU-016. Surprisingly, unlike TRU-016, these antibodies had potent apoptotic activity in the absence of cross-linking agent. After humanization by variable domain re-surfacing, the selected antibodies retained high affinity binding to CD37+ B-cells with an EC50 of < 1 nM. They had much stronger pro-apoptotic activity than rituximab against Ramos cells, with the K7153A antibody among those with the best EC50. They all had antibody-dependent cell-mediated cytotoxicity (ADCC) activity, with K7153A having the most potent activity against Daudi cells. When SMCC-DM1 conjugates of humanized antibodies were compared, the K7153A-SMCC-DM1 conjugate had the most potent specific cytotoxicity against Daudi and Granta-519 cells in vitro. Therefore, the K7153A anti-CD37 antibody provided the best overall anti-tumor activity in terms of its direct pro-apoptotic activity, effector function and potency when used in an AMC. To determine the most effective linker design, maytansinoid conjugates of K7153A were prepared with either hindered disulfide (SPP-DM1) or thioether (SMCC-DM1) linker chemistries. Both conjugates were highly active against lymphoma cells in vitro, with the SMCC-DM1 conjugate being somewhat more potent. In vivo, a single dose of either 10 mg/kg of K7153A-SMCC-DM1 or 5 mg/kg of K7153A-SPP-DM1 was highly active against established SU-DHL-4 sc xenograft tumors. Both treatments resulted in >50% tumor-free survivors at study end. Similarly, the same treatment dose and schedule resulted in good efficacy with both conjugates in a BJAB sc xenograft model. Thus, the K7153A-SMCC-DM1 conjugate was highly active against lymphoma xenograft tumors and, based on preclinical experience, is expected to have comparable, if not better, therapeutic index to that of the SPP-linked conjugate. Taken together, these data support the selection of the K7153A antibody and the SMCC-DM1 design as the optimal anti-CD37 antibody-maytansinoid conjugate for clinical development (designated IMGN529). Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2830. doi:10.1158/1538-7445.AM2011-2830


Cancer Research | 2011

Abstract 4565: IMGN529: A therapeutic maytansinoid conjugate of an anti-CD37 antibody with multiple mechanisms of action for B-cell lymphoma and leukemia

Jutta Deckert; Michele Mayo; Yong Yi; Min Li; Sharon Chicklas; Daniel Tavares; Jan Pinkas; Thomas D. Chittenden; Robert J. Lutz; John M. Lambert; Peter U. Park

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL CD37 is a B-cell surface antigen that is an attractive target for antibody and antibody-drug conjugate mediated therapies due to its restricted expression profile. It is expressed on malignant B-cells in NHL and CLL, but on normal tissue its expression is highly restricted to B-cells present in blood and lymphoid tissues. A large panel of anti-CD37 murine monoclonal antibodies were generated and screened for their specific CD37 binding affinity, direct anti-proliferative activity and pro-apoptotic activity against lymphoma cell lines. Selected antibodies were humanized by variable domain resurfacing and one antibody, designated K7153A, demonstrated the best overall activity in terms of direct antibody activity as well as effector function. K7153A demonstrated much stronger pro-apoptotic activity against Ramos and Raji cells than either of two reference compounds, the anti-CD37 SMIP TRU-016 or the anti-CD20 antibody rituximab, and did not require cross-linking to achieve this effect. The antibody-maytansinoid conjugate, IMGN529, was produced by conjugation of K7153A with the potent maytansinoid, DM1, via the non-cleavable linker, SMCC. IMGN529 retains the high specific binding affinity of the K7153A antibody, with an EC50 of 0.5 nM. IMGN529 also demonstrated the same strong pro-apoptotic activity as the K7153A antibody against Ramos cells, with an EC50 of 0.1 nM. Antibody-dependent cell-mediated cytotoxicity (ADCC) assays, using purified human NK cells as effector cells, showed that K7153A and IMGN529 have similar potent ADCC activity against Ramos and Daudi cells with an EC50 of less than 10 pM. In addition, both K7153A and IMGN529 demonstrated comparable complement-dependent cytotoxicity (CDC) in the presence of human complement against Ramos cells. These results indicate that IMGN529 retains the intrinsic functions of the K7153A antibody. IMGN529 was highly cytotoxic in vitro against NHL cell lines such as Daudi, BJAB, Namalwa and SU-DHL-4 with a greater degree of cell killing and lower EC50 value (19 – 36 pM) than the K7153 antibody alone. In contrast, TRU-016 showed no effect on any of these cell lines and rituximab was only active against SU-DHL-4 cells. In vivo, IMGN529 showed markedly higher efficacy against established SU-DHL-4 and BJAB xenograft tumors than the K7153A antibody alone, with significant anti-tumor activity at single doses of 5 mg/kg or lower. Together, these results demonstrate that IMGN529 combines the strong pro-apoptotic activity, CDC and ADCC activity of its anti-CD37 antibody component with the potent cytotoxic activity provided by the targeted delivery of its maytansinoid payload. IMGN529 is a highly active antibody-drug conjugate with a unique combination of anti-tumor activities and is therefore a promising therapeutic candidate for the treatment of CD37-positive lymphomas and leukemias. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4565. doi:10.1158/1538-7445.AM2011-4565


Cancer Research | 2011

Abstract 4581: IMGN529 is an anti-CD37-maytansinoid conjugate with potent in vitro and in vivo activity against NHL and CLL cell lines

Michele Mayo; Min Li; Yong Yi; Shanqin Xu; Katharine C. Lai; Hans K. Erickson; Jutta Deckert; Peter U. Park; Robert J. Lutz; Jan Pinkas

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL IMGN529 is an antibody-drug conjugate consisting of the functional anti-CD37 antibody K7153A conjugated by a non-cleavable SMCC linker to the highly cytotoxic maytansinoid, DM1. Expression of CD37 in normal tissues is mainly limited to B-cells in blood and lymphoid tissues such as spleen, tonsil and lymph nodes. To evaluate the potential for targeting CD37 in B-cell malignancies by IMGN529, CD37 expression was compared to that of CD20, a well-validated target for antibody-directed therapy in lymphoma. CD37 exhibited a similar prevalence as CD20 in subtypes of NHL such as follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), Burkitts lymphoma (BL) and mantle cell lymphoma (MCL) as assessed by immunohistochemistry using tumor microarrays. In addition, CD37 and CD20 antigen levels were measured by quantitative flow cytometry in NHL and B-CLL cell lines. All of the CD20+ B-cell lines examined also expressed CD37 at antigen levels ranging from 17,000 to 360,000 ABC (antibodies bound per cell). The activity of IMGN529 and its antibody component (K7153A) were evaluated against NHL and CLL cell lines in vitro. K7153A was active against an array of NHL-derived lymphoma cell lines including the DLBCL cell line SU-DHL-4, the FL cell line DOHH-2, the MCL cell line Granta-519, as well as B-CLL cell lines such as JVM-2 and JVM-3. The antibody showed only minimal activity against the BL cell lines Daudi and BJAB. Importantly, IMGN529 demonstrated enhanced and specific cytotoxicity against these cell lines when compared to either unconjugated K7153A antibody or a non-targeted control conjugate (EC50 10 – 400 pM). In contrast, the anti-CD37 compound, TRU-016, had no direct effect on any of these cell lines. The anti-CD20 antibody rituximab showed less direct activity than K7153A in most of these cell lines despite the often higher CD20 expression levels. In vivo, while the K7153A antibody showed anti-tumor efficacy in established tumor xenograft models of DLBCL (SU-DHL-4), FL (DOHH-2) and B-CLL (JVM-3), IMGN529 was significantly more active in these models. In addition, IMGN529 was active against established BL-derived BJAB tumors at doses (5 or 10 mg/kg single dose) where the unconjugated antibody was not active. In the FL-derived DOHH-2 tumor model, a single treatment of IMGN529 at 10 mg/kg resulted in tumor growth delay that was similar to rituximab given at 5 mg/kg twice weekly for 3 weeks and was better than treatment with CVP (cyclophosphamide/vincristine/prednisone). A single 5 mg/kg dose of IMGN529 demonstrated better activity than the anti-CD20 antibody ofatumumab (5 mg/kg twice weekly × 3) or bendamustine (50 mg/kg x1) in the B-CLL-derived JVM-3 xenograft model. In conclusion, IMGN529 is highly active in vitro and in vivo against representative B-cell lines from NHL and CLL suggesting the utility of this functional antibody-maytansinoid conjugate as a therapeutic in these disease indications. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4581. doi:10.1158/1538-7445.AM2011-4581

Collaboration


Dive into the Michele Mayo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge