scholarly journals Targeting the BCL-2-regulated apoptotic pathway for the treatment of solid cancers

Author(s):  
W. Douglas Fairlie ◽  
Erinna F. Lee

The deregulation of apoptosis is a key contributor to tumourigenesis as it can lead to the unwanted survival of rogue cells. Drugs known as the BH3-mimetics targeting the pro-survival members of the BCL-2 protein family to induce apoptosis in cancer cells have achieved clinical success for the treatment of haematological malignancies. However, despite our increasing knowledge of the pro-survival factors mediating the unwanted survival of solid tumour cells, and our growing BH3-mimetics armamentarium, the application of BH3-mimetic therapy in solid cancers has not reached its full potential. This is mainly attributed to the need to identify clinically safe, yet effective, combination strategies to target the multiple pro-survival proteins that typically mediate the survival of solid tumours. In this review, we discuss current and exciting new developments in the field that has the potential to unleash the full power of BH3-mimetic therapy to treat currently recalcitrant solid malignancies.

1976 ◽  
Vol 4 (1) ◽  
pp. 74-78 ◽  
Author(s):  
J Borges ◽  
C Zavaleta

The effect of a new analgesic compound ( propoxyphene, acetaminophen, caffeine, hydroxyzine) was investigated in a single-blind study comparing it with plain acetaminophen administered to forty patients with tension headache. For the study, patients were assigned to one of two groups of twenty each. Starting dose for each group was one to two tablets followed by one tablet every four to six hours. The results show that 90% clinical success was obtained with the analgesic compound, while a 45% success was obtained with plain acetaminophen. This is a statistically significant difference. Side-effects observed with analgesic compound were primarily drowsiness and dizziness of mild intensity; acetaminophen caused gastro-intestinal alterations ( nausea, vomiting) and dizziness of greater severity. Therapy was withdrawn in 20% of patients taking acetaminophen because of side-effects. The dosage of analgesic compound required to control each episode of tension headache way smaller than that of acetaminophen. These results can be explained by a possible potentiation of pharmacological activity of the compound's components. It can be concluded that the analgesic compound is a new and effective combination for the symptomatic treatment of tension headache.


F1000Research ◽  
2016 ◽  
Vol 5 ◽  
pp. 2804 ◽  
Author(s):  
Haiming Dai ◽  
X. Wei Meng ◽  
Scott H. Kaufmann

The BCL2-selective BH3 mimetic venetoclax was recently approved for the treatment of relapsed, chromosome 17p-deleted chronic lymphocytic leukemia (CLL) and is undergoing extensive testing, alone and in combination, in lymphomas, acute leukemias, and solid tumors. Here we summarize recent advances in understanding of the biology of BCL2 family members that shed light on the action of BH3 mimetics, review preclinical and clinical studies leading to the regulatory approval of venetoclax, and discuss future investigation of this new class of antineoplastic agent.


Cancers ◽  
2020 ◽  
Vol 12 (11) ◽  
pp. 3219
Author(s):  
Louise Müller ◽  
Robert Berkeley ◽  
Tyler Barr ◽  
Elizabeth Ilett ◽  
Fiona Errington-Mais

Oncolytic virotherapy (OVT) has received significant attention in recent years, especially since the approval of talimogene Laherparepvec (T-VEC) in 2015 by the Food and Drug administration (FDA). Mechanistic studies of oncolytic viruses (OVs) have revealed that most, if not all, OVs induce direct oncolysis and stimulate innate and adaptive anti-tumour immunity. With the advancement of tumour modelling, allowing characterisation of the effects of tumour microenvironment (TME) components and identification of the cellular mechanisms required for cell death (both direct oncolysis and anti-tumour immune responses), it is clear that a “one size fits all” approach is not applicable to all OVs, or indeed the same OV across different tumour types and disease locations. This article will provide an unbiased review of oncolytic reovirus (clinically formulated as pelareorep), including the molecular and cellular requirements for reovirus oncolysis and anti-tumour immunity, reports of pre-clinical efficacy and its overall clinical trajectory. Moreover, as it is now abundantly clear that the true potential of all OVs, including reovirus, will only be reached upon the development of synergistic combination strategies, reovirus combination therapeutics will be discussed, including the limitations and challenges that remain to harness the full potential of this promising therapeutic agent.


Blood ◽  
2021 ◽  
Author(s):  
Rachel Thijssen ◽  
Sarah T Diepstraten ◽  
Donia M Moujalled ◽  
Edward Chew ◽  
Christoffer Flensburg ◽  
...  

Selective targeting of BCL2 with the BH3-mimetic venetoclax is proving transformative for patients with various leukemias. TP53 controls apoptosis upstream from where BCL2 and its pro-survival relatives, such as MCL1, act. Therefore, targeting these pro-survival proteins could trigger apoptosis across diverse blood cancers, irrespective of TP53 mutation status. Indeed, targeting BCL2 has produced clinically relevant responses in blood cancers with aberrant TP53. However, we show that TP53 mutated or deficient myeloid and lymphoid leukemias outcompete isogenic controls with intact TP53, unless sufficient concentrations of BH3-mimetics targeting BCL2 or MCL1 are applied. Strikingly, tumor cells with TP53 dysfunction escape and thrive over time if inhibition of BCL2 or MCL1 is sub-lethal, in part because of an increased threshold for BAX/BAK activation in these cells. Our study reveals the key role of TP53 in shaping long-term responses to BH3-mimetic drugs and reconciles the disparate pattern of initial clinical response to venetoclax, followed by subsequent treatment failure among patients with TP53-mutant chronic lymphocytic leukemia (CLL) or acute myeloid leukemia (AML). In contrast to BH3-mimetics targeting just BCL2 or MCL1 at doses which are individually sub-lethal, we find that a combined BH3-mimetic approach targeting both pro-survival proteins enhances lethality and durably suppresses leukemic burden, regardless of TP53 mutation status. Our findings highlight the importance of employing sufficiently lethal treatment strategies to maximize outcomes for patients with TP53-mutant disease. In addition, our findings caution against use of sub-lethal BH3-mimetic drug regimens, which may enhance the risk of disease progression driven by emergent TP53 mutant clones.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 567-567
Author(s):  
Donia M Moujalled ◽  
Diane T Hanna ◽  
Giovanna Pomilio ◽  
Veronique Litalien ◽  
Shaun Fleming ◽  
...  

Abstract Background Precursor-B acute lymphoblastic leukemia (B-ALL) is an aggressive hematological malignancy. Relapsed disease has a poor prognosis, despite improved outcomes with tyrosine kinase inhibitors for Ph+ cases and immunotherapeutic approaches, such as blinotumomab and CAR-T cells. Targeting cell survival with novel small molecule BH3-mimetic inhibitors of BCL-2 (e.g. Souers et al Nat Med 2013, Roberts et al, NEJM 2016 and Casara et al, Oncotarget 2018), BCL-XL (Lessene et al, Nat Chem Biol, 2013) or MCL1 (Kotschy et al, Nature 2016) is an emerging therapeutic option. BCL-2 is reported to have a pro-survival role in BCR-ABL1, JAK2 fusion, ETV6-RUNX1 and MLL-r driven ALL (Brown et al., Journal Biological Chemistry 2017). BH3-mimetics targeting BCL-2 and BCL-XL has efficacy in paediatric ALL xenografts (Khaw et al., Blood 2016), while ruxolitinib combined with ABT-737 is synergistic in JAK2-mutant pre-B-ALL (Waibel et al., Cell Reports 2013). We now report that combined targeting of BCL-2 and MCL1 has broad pre-clinical efficacy in adult B-ALL samples with Ph+, Ph- and Ph-like characteristics. Methods S55746 and S63845 were obtained from Servier/Novartis, A1331852 from Guillaume Lessene (WEHI), venetoclax, daunorubicin, dexamethasone (DXM) and tyrosine kinase inhibitors (TKIs) from Selleckchem. Bliss synergy scores were determined using a checkerboard approach to evaluate combinations (previously described Bliss, Ann Appl Biol 1939). Primary ALL cells were obtained from 14 patients (4 Ph+ and 10 Ph-) providing informed consent. Ex vivo cell viability (sytox blue exclusion) at 48h was determined over a 5-log dilution range (1nM-10uM) using drugs alone or in equimolar combinations. For in vivo studies, adult B-ALL patient derived xenografts were performed in NSG; NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice. Results Dual BH3-mimetic targeting of BCL-2 and MCL1 was strongly synergistic (Bliss sum >1000) in SUPB15 (Ph+ BCR-ABL1), BV173 (lymphoid blast crisis BCR-ABL1), MUTZ5 (Ph-like) and MHHCALL4 (Ph-like) B-ALL cell lines. This was more effective than single BH3-mimetic combinations with DXM or TKIs (dasatinib or ruxolitinib) (Fig. A, B). In B-ALL patient samples, combined BCL-2 and MCL1 targeting lowered the LC50 by 10-1000 fold (to LC50<10nM) in 4/4 Ph+ ALL cases and 8/10 Ph- cases. Similarly, combined MCL1 and BCL-XL targeting demonstrated synergy in 3/4 Ph+ cases and 7/10 Ph- cases (to LC50<10nM), confirming remarkable anti-leukemic activity compared to BH3-mimetics alone or chemotherapy (daunorubicin) (Fig. C). BH3-mimetic combination therapy (S55746/S63845) compared favourably in Ph+ ALL cases to S55746 (figure D) or S63845 (Figure E) in combination with dasatinib. Preliminary data using patient-derived xenografts in NSG mice revealed in vivo efficacy of combined S55746 and S63845 therapy against 3 adult B-ALL cases (1 Ph+ and 2 Ph-). Reduction of established ALL in the bone marrow was observed in mice receiving combined S55746/S63845 after one week of treatment (p=<0.05) (Fig. F-H). Conclusions Dual BH3-mimetic targeting of BCL-2 and MCL1 induces synergistic killing of human B-ALL cell lines and primary ALL samples in vitro and rapid cytoreduction in vivo. Simultaneous inhibition of BCL-2 and MCL1 represents a novel and effective approach for targeting Ph+, Ph- and Ph-like B-ALL without need for additional DNA-damaging chemotherapy or kinase inhibition. Our results support the translational investigation of dual BH3-mimetic targeting of BCL-2 and MCL1 in the clinic. Figure legend: BLISS synergy scores for A. Ph+ and B. Ph-like ALL cell lines for drug combinations targeting BCL-2, MCL1, BCR-ABL, JAK1/2 and DXM. C. LC50 activity in primary ALL after 48hr of treatment with BH3 mimetics and combinations targeting BCL-2, MCL1, BCL-XL, compared to daunorubicin (LC50< 10nM red; ~ 100nM yellow; >1uM green). D. Comparison of BH3-mimetics targeting D.BCL-2 or E. MCL1 in combination with dasatinib in Ph+ vs Ph- primary B-ALL samples. Activity expressed as LC50 activity after 48h, with median values shown. Irradiated NSG mice were transplanted with 106 primary B-ALL cells. Engraftment of F. Ph+ and G-H. Ph- B-ALL cells was confirmed at 10 weeks by detection of hCD45 in PB. Mice were then treated with i) vehicle (d1-5), ii) S55746 100mg/kg days 1-5 by gavage, iii) S63845 25 mg/kg IV on days 2 and 4 or iv) S55746+S63845. Mice were euthanized on day 8 and hCD45+ from flushed femurs quantified. Disclosures Chanrion: Servier: Employment. Maragno:servier: Employment. Kraus-Berthier:servier: Employment. Lessene:servier: Research Funding. Roberts:Janssen: Research Funding; AbbVie: Research Funding; Genentech: Research Funding; Walter and Eliza Hall: Employment, Patents & Royalties: Employee of Walter and Eliza Hall Institute of Medical Research which receives milestone and royalty payments related to venetoclax. Geneste:servier: Employment. Wei:Pfizer: Honoraria, Other: Advisory committee; Celgene: Honoraria, Other: Advisory committee, Research Funding; Amgen: Honoraria, Other: Advisory committee, Research Funding; Servier: Consultancy, Honoraria, Other: Advisory committee, Research Funding; Novartis: Honoraria, Other: Advisory committee, Research Funding, Speakers Bureau; Abbvie: Honoraria, Other: Advisory board, Research Funding, Speakers Bureau.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 14-15
Author(s):  
Valentina Cordo' ◽  
Rico Hagelaar ◽  
Sander Piersma ◽  
Richard Goeij-de Haas ◽  
Thang V Pham ◽  
...  

Introduction Intensive multi-agent treatment has boosted survival up to 80% of pediatric T-cell acute lymphoblastic leukemia (T-ALL) patients. Nevertheless, relapsed patients have a poor prognosis due to acquired therapy resistance while most survivors have detrimental chemotherapy-induced side effects. Therefore, novel targeted therapies are urgently needed since further intensification of the current standard treatment regimen is not feasible for refractory/relapsed cases. Protein kinase inhibitors (PKIs) are amongst the most successful cancer treatments. Targetable kinases activated by gene fusions are rare in T-ALL and include subclonal NUP214-ABL1 fusion in 6% of cases or other rare clonal ABL1 fusions. Nevertheless, leukemic blasts rely on enhanced kinase signaling to sustain their dysregulated proliferation. Protein kinases can be hyper-activated even in the absence of defects in their genes. Thus, together with the identification of genomic aberrations, phospho-proteomics can provide information on pathway activation, signaling networks and aberrant kinase activities that offer important opportunities for targeted therapies. Aim Here, we aimed to identify and quantify kinase activation in T-ALL cell lines that may yield differential sensitivity to PKIs in vitro. This approach could pinpoint targetable leukemia vulnerabilities and provide effective (combination) treatment strategies. Methods Protein extracts from 11 T-ALL cell lines were enriched for phospho-peptides by titanium dioxide enrichment and anti-phospho tyrosine immunoprecipitation followed by liquid chromatography - tandem mass spectrometry (MS). Subsequently, the Integrative Inferred Kinase Activity (INKA) pipeline was used to rank activated kinases in our panel (Beekhof et al., 2019). Based on these data, selected kinase inhibitors were tested in vitro as single treatment or in combinations. Eventually, drugs of interest were further tested ex vivo in a cohort of T-ALL patient-derived xenografts (PDXs). Results MS-based phospho-proteome profiling of 11 T-ALL cell lines identified about 3700 tyrosine phospho-sites and more than 13300 serine/threonine phosphorylation sites. We found SRC-family kinases including LCK, SRC, FYN, and YES1 as most activated kinases in many T-ALL cell lines while ABL1, ZAP70, LYN, and FGR were detected only in specific lines. Additionally, other kinases including CDK1/2 and PAK1/2 were found to be activated in all the cell lines while activation of the INSR/IGF-1R axis was detected only in a subset of lines. We then tested cellular response to multiple clinically relevant PKIs based on predicted kinase activities. The in vitro drug screening showed an effective response and G1-arrest following treatment with the CDK1/2 inhibitor milciclib in all the cell lines tested, with IC50 values between 10nM and 1uM. Despite a general SRC-family kinases activation profile, dual SRC/ABL inhibitors like dasatinib reduced cellular viability only in cell lines with ABL1 fusions or LCK translocations (IC50 &lt; 10nM) while other lines lacking ABL or LCK rearrangements were affected at much higher drug concentration (IC50 &gt; 3uM, which is beyond the clinical achievable plasma concentration). Thus, PKIs were tested in combination with other relevant inhibitors based on additional kinase activities detected. Interestingly, the concomitant inhibition of the SRC-family kinases by dasatinib and the INSR/IGF-1R axis by BMS-754807 led to a drastic reduction of cell viability at nanomolar concentrations even in cell lines that did not respond to dasatinib, identifying a novel possible effective combination strategy for T-ALL. Eventually, we tested clinically relevant PKIs in 50 PDXs ex vivo and identified various T-ALL samples with a high sensitivity to dasatinib single treatment (IC50 &lt; 100nM) as previously reported by others (Frismantas et al., 2017). Moreover, 70% of our PDXs efficiently responded to the broad spectrum kinase inhibitor midostaurin (IC50 &lt; 1uM), highlighting the importance of targeting multiple signaling nodes simultaneously to tackle T-ALL vulnerabilities. Conclusions Ranking kinase activities and signaling networks from phospho-proteomic data can guide the use of PKIs as treatment option for T-ALL patients. Moreover, kinase activity profiling can provide insights for efficient treatment combination strategies to develop personalized medicine. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
◽  
Kaitlyn Smith

Anti-apoptotic MCL1 is one of the most frequently amplified genes in human cancers and its elevated expression confers resistance to many therapeutics including the BH3-mimetic agents ABT-199 and ABT-263. The anti-malarial, dihydroartemisinin (DHA) translationally represses MCL-1 and synergizes with BH3-mimetics. To explore how DHA represses MCL-1, a genome-wide CRISPR screen identified that loss of genes in the heme synthesis pathway renders mouse BCR-ABL+ B-ALL cells resistant to DHA-induced death. Mechanistically, DHA disrupts the interaction between heme and the eIF2α kinase heme regulated inhibitor (HRI) triggering the integrated stress response. Genetic ablation of Eif2ak1, which encodes HRI, blocks MCL-1 repression in response to DHA treatment and represses the synergistic killing of DHA and BH3-mimetics compared to wild-type leukemia. Furthermore, BTdCPU, a small-molecule activator of HRI, similarly triggers MCL-1 repression and synergizes with BH3-mimetics in mouse and human leukemia including both Ph+ and Ph-like B-ALL. Lastly, combinatorial treatment of leukemia bearing mice with both BTdCPU and a BH3-mimetic extended survival and repressed MCL-1 in vivo. These findings reveal that the HRI-dependent cellular heme-sensing pathway can modulate apoptosis in leukemic cells by repressing MCL-1 and increasing their responsiveness to BH3-mimetics. This signaling pathway could represent a generalizable mechanism for repressing MCL-1 expression in malignant cells and sensitizing them to available therapeutics.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1486-1486
Author(s):  
Andrew W. Roberts ◽  
Andrew Wei ◽  
Mark F. van Delft ◽  
Kylie D. Mason ◽  
Jerry M. Adams ◽  
...  

Abstract As overactivity of pro-survival Bcl-2 proteins promotes neoplasia and enhances resistance of malignant cells to cytotoxic therapies, a promising approach for incurable lymphoid tumors is to directly target the pro-survival proteins. One approach is to mimic their physiological antagonists, the BH3-only proteins. We initially determined which of the diverse BH3-only proteins would be optimal to mimic. The interactions of the BH3 domains of this family with a groove on the Bcl-2-like proteins have been considered promiscuous. However, we found that the interactions between eight BH3 peptides and five Bcl-2-like proteins varied over 10,000 fold in affinity, and that only certain protein pairs associate inside cells (Chen et al Mol Cell;17:393–403, 2005). Bim and Puma potently engaged all the pro-survival proteins comparably. Bad, however, bound preferentially to Bcl-2, Bcl-xL and Bcl-w. Strikingly, Noxa bound only Mcl-1 and A1. In accord with their complementary binding, Bad and Noxa cooperated to induce potent killing. We next determined the mechanism of action of putative BH3 mimetic compounds. Because killing by the BH3-only proteins require the action of the essential cell death mediators, Bax and Bak, we initially screened putative BH3 mimetic compounds on cells genetically engineered to lack both Bax and Bak. Only those compounds that mimicked the BH3-only proteins (i.e, inert on BaxBak double null cells, but active in Bax or Bak expressing cells) were evaluated further. Surprisingly, among seven putative BH3 mimetics tested, we found that only the recently described ABT-737 (Abbott Laboratories; Oltersdorf et al, Nature435:677–81, 2005), required the pro-apoptotic protein Bax or Bak to induce apoptosis. The cytotoxicity of ABT-737 alone was modest in a range of hematopoietic and non-hematopoietic cell types. Further investigations revealed that, like Bad, ABT-737 only targeted Bcl-2, Bcl-xL and Bcl-w. As hematopoietic cells typically express Mcl-1, we attempted to augment the activity of ABT-737 by concomitantly neutralizing Mcl-1. Targeting of Mcl-1 by overexpressing the BH3-only protein Noxa, or Mcl-1 down-regulation by RNAi, cytokine deprivation or cytotoxic agents, allowed ABT-737 to efficiently kill diverse cell types, even when Bcl-2 was over-expressed. We conclude that ABT-737 is a highly selective and specific BH3 mimetic compound that should prove highly efficacious in tumors where Mcl-1 is low, or when combined with agents that down-regulate Mcl-1. This hypothesis is currently being tested in vitro in primary human lymphoid malignancies, and in vivo using immunocompetent murine models of lymphoma. ABT-737 also provides strong proof-of-principle that targeting pro-survival Bcl-2 proteins is feasible, but the optimal utility of such BH3 mimetics depends on a thorough understanding of the pathways to apoptosis.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3293-3293
Author(s):  
Jia-Nan Gong ◽  
Tiffany Khong ◽  
David Segal ◽  
Yuan Yao ◽  
Mark F van Delft ◽  
...  

Abstract Multiple myeloma is a clonal B-cell malignancy characterized by complex genetic aberrations. Despite recent advances in our understanding of genetic basis of this disease and the introduction of novel therapies, the outcome for many patients remains poor. For example, 20% of newly diagnosed patients with stage 3 myeloma have disease marked by high lactate dehydrogenase levels or unfavorable cytogenetic abnormalities (e.g. 17p deletion, t(4;14)). Half of these patients die within 2 years of diagnosis in spite of advanced therapies. A promising class of novel agents to treat patients with other B-cell malignancies is the BH3 mimetic compounds, which targets the pro-survival BCL2 proteins. For example, the majority of patients with CLL (chronic lymphocytic leukaemia) respond to venetoclax (ABT-199), a BCL2-selective inhibitor. Myeloma cell lines with t(11;14) translocations are also reported to be sensitive to BCL2 inhibition and venetoclax is in clinic trails for patients with relapsed or refractory multiple myeloma. We are eager to know whether the other pro-survival BCL2 proteins, namely BCLXL, BCLW, MCL1 and BCL2A1 (BFL1) play a role in maintaining the survival of myeloma cells. To determine this, we screened a large panel of immortalized (25) and low-passage (7) myeloma cell lines to killing by BH3 mimetics with varying specificities. We found that a fraction of the myeloma cell lines were rapidly killed when BCL2 (25%, 8/32) or BCLXL (25%, 8/32) were targeted. Interestingly, we also identified a distinct t(4;14) subgroup, as well as the previous recognized t(11;14) subgroup, to be highly sensitive to BCL2 inhibition. Unlike CLL, in which the majority patients respond to venetoclax, BCL2 inhibition is only likely to account for some cases. We then asked which other pro-survival BCL2 protein keeps the other myeloma cell lines alive. A prime candidate is MCL1 since normal plasma cells rely on it and previous studies had implicated MCL1 in myeloma. By targeting the expression of the pro-survival BCL2 proteins using CRISPR/Cas9 genome editing technology, we found that a significant fraction (74%, 14/19) of our cell line panel died rapidly in the absence of MCL1. The importance of MCL1 was confirmed using an orthogonal approach: we found that expression of a MCL1-selective peptidyl ligand BIM2A rapidly killed a sizeable fraction of both immortalized (17/25) and low-passage (5/7) myeloma cell lines. This activity strongly correlated with the genetic targeting of MCL1 by CRISPR/Cas9. Importantly, even cell lines that harbor the poor prognostic t(4;14) chromosomal translocation were readily killed by BIM2A and the cell lines were killed regardless of their TP53 status. Moreover, targeting MCL1 also constrained the growth of myeloma in vivo. Since the inhibition of MCL1, BCLXL or BCL2 can on their own can kill almost all the myeloma cell lines screened, we next identified the molecular mechanisms by which these BH3 mimetics act. Firstly, we tested likely candidates for their role in driving the killing induced by a specific BH3 mimetic, for example whether deleting the BH3-only protein BIM impact upon killing by venetoclax. Secondly, we undertook genome-wide recessive CRISPR/Cas9 screens for genes that are critical for the action of the BH3 mimetics. We have identified a number of hits from such screens and are in the process of validating these. By identifying the unique susceptibility of the myeloma cell lines to the BH3 mimetics and elucidating how they act to kill, our studies are critical for the clinical testing of BH3 mimetics that target BCL2 or its close pro-survival relatives in patients with multiple myeloma. Disclosures Gong: The Walter and Eliza Hall Institute of Medical Research: Employment. Segal:The Walter and Eliza Hall Institute of Medical Research: Employment. van Delft:The Walter and Eliza Hall Institute of Medical Research: Employment. Roberts:The Walter and Eliza Hall Institute of Medical Research: Employment. Huang:The Walter and Eliza Hall Institute of Medical Research: Employment.


Sign in / Sign up

Export Citation Format

Share Document