Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cureus ; 16(4): e59052, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38800313

ABSTRACT

Malignant mesothelioma of the tunica vaginalis testis (MMTVT) is a rare tumor of the testicular sheath. More than 50% of cases initially present as recurrent hydrocele, but there have also been documented cases with hematocele, inguinal hernia, or epididymitis. Due to the non-specific symptoms and signs of the disease, it is almost always diagnosed intra- or postoperatively. The lack of standardized therapy protocols, as well as the lack of evidence supporting systemic chemotherapy, have fueled arguments in favor of the necessity of retroperitoneal lymph node dissection (RPLND) in the treatment of the disease, especially in the case of lymph node metastasis. We present a case of MMTVT achieving prolonged remission after timely and extensive surgical treatment.

2.
JAMA Oncol ; 8(4): 546-552, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35142815

ABSTRACT

IMPORTANCE: DCVAC/PCa is an active cellular immunotherapy designed to initiate an immune response against prostate cancer. OBJECTIVE: To evaluate the efficacy and safety of DCVAC/PCa plus chemotherapy followed by DCVAC/PCa maintenance treatment in patients with metastatic castration-resistant prostate cancer (mCRPC). DESIGN, SETTING, AND PARTICIPANTS: The VIABLE double-blind, parallel-group, placebo-controlled, phase 3 randomized clinical trial enrolled patients with mCRPC among 177 hospital clinics in the US and Europe between June 2014 and November 2017. Data analyses were performed from December 2019 to July 2020. INTERVENTIONS: Eligible patients were randomized (2:1) to receive DCVAC/PCa (add-on and maintenance) or placebo, both in combination with chemotherapy (docetaxel plus prednisone). The stratification was applied according to geographical region (US or non-US), prior therapy (abiraterone, enzalutamide, or neither), and Eastern Cooperative Oncology Group performance status (0-1 or 2). DCVAC/PCa or placebo was administered subcutaneously every 3 to 4 weeks (up to 15 doses). MAIN OUTCOMES AND MEASURES: The primary outcome was overall survival (OS), defined as the time from randomization until death due to any cause, in all randomized patients. Survival was compared using 2-sided log-rank test stratified by geographical region, prior therapy with abiraterone and/or enzalutamide, and Eastern Cooperative Oncology Group performance status. RESULTS: A total of 1182 men with mCRPC (median [range] age, 68 [46-89] years) were randomized to receive DCVAC/PCa (n = 787) or placebo (n = 395). Of these, 610 (81.8%) started DCVAC/PCa, and 376 (98.4%) started placebo. There was no difference in OS between the DCVAC/PCa and placebo groups in all randomized patients (median OS, 23.9 months [95% CI, 21.6-25.3] vs 24.3 months [95% CI, 22.6-26.0]; hazard ratio, 1.04; 95% CI, 0.90-1.21; P = .60). No differences in the secondary efficacy end points (radiological progression-free survival, time to prostate-specific antigen progression, or skeletal-related events) were observed. Treatment-emergent adverse events related to DCVAC/PCa or placebo occurred in 69 of 749 (9.2%) and 48 of 379 (12.7%) patients, respectively. The most common treatment-emergent adverse events (DCVAC/PCa [n = 749] vs placebo [n = 379]) were fatigue (271 [36.2%] vs 152 [40.1%]), alopecia (222 [29.6%] vs 130 [34.3%]), and diarrhea (206 [27.5%] vs 117 [30.9%]). CONCLUSIONS AND RELEVANCE: In this phase 3 randomized clinical trial, DCVAC/PCa combined with docetaxel plus prednisone and continued as maintenance treatment did not extend OS in patients with mCRPC and was well tolerated. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT02111577.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dendritic Cells/pathology , Docetaxel/adverse effects , Double-Blind Method , Female , Humans , Immunotherapy/adverse effects , Male , Prednisone , Prostatic Neoplasms, Castration-Resistant/drug therapy
3.
J Neurotrauma ; 34(3): 685-694, 2017 02.
Article in English | MEDLINE | ID: mdl-27503053

ABSTRACT

Magnesium (Mg2+) homeostasis is impaired following spinal cord injury (SCI) and the loss of extracellular Mg2+ contributes to secondary injury by various mechanisms, including glutamate neurotoxicity. The neuroprotective effects of high dose Mg2+ supplementation have been reported in many animal models. Recent studies found that lower Mg2+ doses also improved neurologic outcomes when Mg2+ was formulated with polyethylene glycol (PEG), suggesting that a PEG/ Mg2+ formulation might increase Mg2+ delivery to the injured spinal cord, compared with that of MgSO4 alone. Here, we assessed spinal extracellular Mg2+ and glutamate levels following SCI in rats using microdialysis. Basal levels of extracellular Mg2+ (∼0.5 mM) were significantly reduced to 0.15 mM in the core and 0.12 mM in the rostral peri-lesion area after SCI. A single intravenous infusion of saline or of MgSO4 at 192 µmoL/kg did not significantly change extracellular Mg2+ concentrations. However, a single infusion of AC105 (a MgCl2 in PEG) at an equimolar Mg2+ dose significantly increased the Mg2+ concentration to 0.3 mM (core area) and 0.25 mM (rostral peri-lesion area). Moreover, multiple AC105 treatments completely restored the depleted extracellular Mg2+ concentrations after SCI to levels in the uninjured spinal cord. Repeated MgSO4 infusions slightly increased the Mg2+ concentrations while saline infusion had no effect. In addition, AC105 treatment significantly reduced extracellular glutamate levels in the lesion center after SCI. These results indicate that intravenous infusion of PEG-formulated Mg2+ normalized the Mg2+ homeostasis following SCI and reduced potentially neurotoxic glutamate levels, consistent with a neuroprotective mechanism of blocking excitotoxicity.


Subject(s)
Drug Delivery Systems/methods , Extracellular Fluid/metabolism , Glutamic Acid/metabolism , Magnesium Sulfate/administration & dosage , Polyethylene Glycols/administration & dosage , Spinal Cord Injuries/metabolism , Animals , Excitatory Amino Acid Agonists , Extracellular Fluid/drug effects , Female , Infusions, Intravenous , Magnesium Sulfate/metabolism , Microdialysis/methods , Polyethylene Glycols/metabolism , Rats , Rats, Long-Evans , Spinal Cord Injuries/drug therapy , Thoracic Vertebrae
4.
Bioorg Med Chem ; 22(15): 4001-9, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24997575

ABSTRACT

The field of small-molecule inhibitors of protein-protein interactions is rapidly advancing and the specific area of inhibitors of the p53/MDM2 interaction is a prime example. Several groups have published on this topic and multiple compounds are in various stages of clinical development. Building on the strength of the discovery of RG7112, a Nutlin imidazoline-based compound, and RG7388, a pyrrolidine-based compound, we have developed additional scaffolds that provide opportunities for future development. Here, we report the discovery and optimization of a highly potent and selective series of spiroindolinone small-molecule MDM2 inhibitors, culminating in RO8994.


Subject(s)
Indoles/chemistry , Indolizidines/chemistry , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Spiro Compounds/chemistry , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Humans , Imidazolines/chemistry , Indoles/therapeutic use , Indoles/toxicity , Indolizidines/therapeutic use , Indolizidines/toxicity , Molecular Dynamics Simulation , Neoplasms/drug therapy , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2/metabolism , Pyrrolidines/chemistry , Spiro Compounds/therapeutic use , Spiro Compounds/toxicity , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , para-Aminobenzoates/chemistry
5.
ACS Med Chem Lett ; 5(2): 124-7, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24900784

ABSTRACT

The development of small-molecule MDM2 inhibitors to restore dysfunctional p53 activities represents a novel approach for cancer treatment. In a previous communication, the efforts leading to the identification of a non-imidazoline MDM2 inhibitor, RG7388, was disclosed and revealed the desirable in vitro and in vivo pharmacological properties that this class of pyrrolidine-based inhibitors possesses. Given this richness and the critical need for a wide variety of chemical structures to ensure success in the clinic, research was expanded to evaluate additional derivatives. Here we report two new potent, selective, and orally active p53-MDM2 antagonists, RO5353 and RO2468, as follow-ups with promising potential for clinical development.

6.
Clin Cancer Res ; 20(14): 3742-52, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24812409

ABSTRACT

PURPOSE: Antitumor clinical activity has been demonstrated for the MDM2 antagonist RG7112, but patient tolerability for the necessary daily dosing was poor. Here, utilizing RG7388, a second-generation nutlin with superior selectivity and potency, we determine the feasibility of intermittent dosing to guide the selection of initial phase I scheduling regimens. EXPERIMENTAL DESIGN: A pharmacokinetic-pharmacodynamic (PKPD) model was developed on the basis of preclinical data to determine alternative dosing schedule requirements for optimal RG7388-induced antitumor activity. This PKPD model was used to investigate the pharmacokinetics of RG7388 linked to the time-course of the antitumor effect in an osteosarcoma xenograft model in mice. These data were used to prospectively predict intermittent and continuous dosing regimens, resulting in tumor stasis in the same model system. RESULTS: RG7388-induced apoptosis was delayed relative to drug exposure with continuous treatment not required. In initial efficacy testing, daily dosing at 30 mg/kg and twice a week dosing at 50 mg/kg of RG7388 were statistically equivalent in our tumor model. In addition, weekly dosing of 50 mg/kg was equivalent to 10 mg/kg given daily. The implementation of modeling and simulation on these data suggested several possible intermittent clinical dosing schedules. Further preclinical analyses confirmed these schedules as viable options. CONCLUSION: Besides chronic administration, antitumor activity can be achieved with intermittent schedules of RG7388, as predicted through modeling and simulation. These alternative regimens may potentially ameliorate tolerability issues seen with chronic administration of RG7112, while providing clinical benefit. Thus, both weekly (qw) and daily for five days (5 d on/23 off, qd) schedules were selected for RG7388 clinical testing.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Bone Neoplasms/drug therapy , Imidazolines/pharmacokinetics , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Drug Administration Schedule , Female , Humans , Imidazolines/therapeutic use , Mice, Nude , Osteosarcoma/drug therapy , Pyrrolidines/pharmacology , Xenograft Model Antitumor Assays , para-Aminobenzoates/pharmacology
7.
Proc Natl Acad Sci U S A ; 110(36): E3445-54, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23946421

ABSTRACT

Stapled α-helical peptides have emerged as a promising new modality for a wide range of therapeutic targets. Here, we report a potent and selective dual inhibitor of MDM2 and MDMX, ATSP-7041, which effectively activates the p53 pathway in tumors in vitro and in vivo. Specifically, ATSP-7041 binds both MDM2 and MDMX with nanomolar affinities, shows submicromolar cellular activities in cancer cell lines in the presence of serum, and demonstrates highly specific, on-target mechanism of action. A high resolution (1.7-Å) X-ray crystal structure reveals its molecular interactions with the target protein MDMX, including multiple contacts with key amino acids as well as a role for the hydrocarbon staple itself in target engagement. Most importantly, ATSP-7041 demonstrates robust p53-dependent tumor growth suppression in MDM2/MDMX-overexpressing xenograft cancer models, with a high correlation to on-target pharmacodynamic activity, and possesses favorable pharmacokinetic and tissue distribution properties. Overall, ATSP-7041 demonstrates in vitro and in vivo proof-of-concept that stapled peptides can be developed as therapeutically relevant inhibitors of protein-protein interaction and may offer a viable modality for cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Peptides/therapeutic use , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Binding, Competitive , Cell Line, Tumor , Crystallography, X-Ray , Female , HCT116 Cells , Humans , MCF-7 Cells , Male , Mice , Mice, Nude , Models, Molecular , Neoplasms/metabolism , Neoplasms/pathology , Peptides/chemistry , Peptides/metabolism , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/therapeutic use , Protein Binding , Protein Conformation , Protein Structure, Secondary , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Rats , Rats, Long-Evans , Xenograft Model Antitumor Assays
8.
J Med Chem ; 56(14): 5979-83, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23808545

ABSTRACT

Restoration of p53 activity by inhibition of the p53-MDM2 interaction has been considered an attractive approach for cancer treatment. However, the hydrophobic protein-protein interaction surface represents a significant challenge for the development of small-molecule inhibitors with desirable pharmacological profiles. RG7112 was the first small-molecule p53-MDM2 inhibitor in clinical development. Here, we report the discovery and characterization of a second generation clinical MDM2 inhibitor, RG7388, with superior potency and selectivity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyrrolidines/chemical synthesis , Tumor Suppressor Protein p53/antagonists & inhibitors , para-Aminobenzoates/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Discovery , Humans , Male , Mice , Mice, Inbred C57BL , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , para-Aminobenzoates/pharmacology , para-Aminobenzoates/therapeutic use
9.
Cancer Res ; 73(8): 2587-97, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23400593

ABSTRACT

MDM2 negatively regulates p53 stability and many human tumors overproduce MDM2 as a mechanism to restrict p53 function. Thus, inhibitors of p53-MDM2 binding that can reactivate p53 in cancer cells may offer an effective approach for cancer therapy. RG7112 is a potent and selective member of the nutlin family of MDM2 antagonists currently in phase I clinical studies. RG7112 binds MDM2 with high affinity (K(D) ~ 11 nmol/L), blocking its interactions with p53 in vitro. A crystal structure of the RG7112-MDM2 complex revealed that the small molecule binds in the p53 pocket of MDM2, mimicking the interactions of critical p53 amino acid residues. Treatment of cancer cells expressing wild-type p53 with RG7112 activated the p53 pathway, leading to cell-cycle arrest and apoptosis. RG7112 showed potent antitumor activity against a panel of solid tumor cell lines. However, its apoptotic activity varied widely with the best response observed in osteosarcoma cells with MDM2 gene amplification. Interestingly, inhibition of caspase activity did not change the kinetics of p53-induced cell death. Oral administration of RG7112 to human xenograft-bearing mice at nontoxic concentrations caused dose-dependent changes in proliferation/apoptosis biomarkers as well as tumor inhibition and regression. Notably, RG7112 was highly synergistic with androgen deprivation in LNCaP xenograft tumors. Our findings offer a preclinical proof-of-concept that RG7112 is effective in treatment of solid tumors expressing wild-type p53.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazolines/pharmacology , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Imidazolines/chemistry , Mice , Molecular Docking Simulation , Neoplasms/pathology , Protein Binding/drug effects , Protein Stability/drug effects , Proto-Oncogene Proteins c-mdm2/chemistry , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Burden/drug effects , Tumor Suppressor Protein p53/chemistry , Xenograft Model Antitumor Assays
10.
Proc Natl Acad Sci U S A ; 103(6): 1888-93, 2006 Feb 07.
Article in English | MEDLINE | ID: mdl-16443686

ABSTRACT

The p53 tumor suppressor retains its wild-type conformation and transcriptional activity in half of all human tumors, and its activation may offer a therapeutic benefit. However, p53 function could be compromised by defective signaling in the p53 pathway. Using a small-molecule MDM2 antagonist, nutlin-3, to probe downstream p53 signaling we find that the cell-cycle arrest function of the p53 pathway is preserved in multiple tumor-derived cell lines expressing wild-type p53, but many have a reduced ability to undergo p53-dependent apoptosis. Gene array analysis revealed attenuated expression of multiple apoptosis-related genes. Cancer cells with mdm2 gene amplification were most sensitive to nutlin-3 in vitro and in vivo, suggesting that MDM2 overexpression may be the only abnormality in the p53 pathway of these cells. Nutlin-3 also showed good efficacy against tumors with normal MDM2 expression, suggesting that many of the patients with wild-type p53 tumors may benefit from antagonists of the p53-MDM2 interaction.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Animals , Cell Cycle/drug effects , Cell Line , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Xenograft Model Antitumor Assays
11.
Science ; 303(5659): 844-8, 2004 Feb 06.
Article in English | MEDLINE | ID: mdl-14704432

ABSTRACT

MDM2 binds the p53 tumor suppressor protein with high affinity and negatively modulates its transcriptional activity and stability. Overexpression of MDM2, found in many human tumors, effectively impairs p53 function. Inhibition of MDM2-p53 interaction can stabilize p53 and may offer a novel strategy for cancer therapy. Here, we identify potent and selective small-molecule antagonists of MDM2 and confirm their mode of action through the crystal structures of complexes. These compounds bind MDM2 in the p53-binding pocket and activate the p53 pathway in cancer cells, leading to cell cycle arrest, apoptosis, and growth inhibition of human tumor xenografts in nude mice.


Subject(s)
Apoptosis/drug effects , Cell Division/drug effects , Imidazoles/pharmacology , Neoplasms, Experimental/pathology , Nuclear Proteins , Piperazines/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Binding Sites , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Crystallization , Crystallography, X-Ray , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Dose-Response Relationship, Drug , Gene Expression , Genes, p53 , Humans , Hydrophobic and Hydrophilic Interactions , Imidazoles/chemistry , Imidazoles/metabolism , Mice , Mice, Nude , Models, Molecular , Molecular Weight , NIH 3T3 Cells , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Phosphorylation , Piperazines/chemistry , Piperazines/metabolism , Protein Conformation , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Stereoisomerism , Transplantation, Heterologous
12.
Mol Cancer Ther ; 2(10): 1023-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14578467

ABSTRACT

The p53 tumor suppressor protein plays a key function in the cellular response to stress by activating a subset of genes responsible for cell cycle arrest and apoptosis. Activation of the p53 pathway in tumor cells has been proposed as a novel approach to cancer therapy and substantial efforts have been dedicated to the discovery of pharmacological p53 activators. Here, we show that the transforming growth factor-beta superfamily cytokine, macrophage inhibitory cytokine-1 (MIC-1), can serve as a secreted biomarker for activation of p53 in both cellular and xenograft models of human cancer. Using doxorubicin treatment in the HCT116 colon cancer cell line, we have shown that MIC-1 secretion into culture media is correlated with p53 pathway activation as measured by the up-regulation of its downstream transcriptional target p21. When transplanted into nude mice, HCT116 cells continued to secret human MIC-1 and mouse plasma levels correlated well with tumor volume. Treatment of these animals with a single dose of doxorubicin led to activation of the p53 pathway and a nearly 4-fold elevation of the plasma MIC-1 level, which was paralleled by p21 induction in the tumor xenografts. Estimation of MIC-1 concentration, both in vivo and in vitro, represents a novel tool for the study of p53 pathway and development of p53-activating therapeutics.


Subject(s)
Cytokines/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Dose-Response Relationship, Drug , Growth Differentiation Factor 15 , Humans , Kinetics , Mice , Mice, Nude , Neoplasm Transplantation , Polymerase Chain Reaction , Time Factors , Tumor Suppressor Protein p53/metabolism
13.
Intensive Care Med ; 28(8): 1164-7, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12185443

ABSTRACT

OBJECTIVE: To examine the short-term cardiovascular effects of plasmapheresis in seven patients (six surgical, one medical) with norepinephrine-refractory septic shock. DESIGN AND SETTING: Retrospective observational study in a secondary community hospital. INTERVENTIONS: Plasmapheresis was performed as follows: blood flow, 120 ml/min; exchange volume, 10 fresh frozen plasma (ca. 2200 ml) at rate of 13 ml/min; anticoagulation, unfractionated heparin 200-300/IU/h. Five patients received one separation and two patients three separations. All patients received norepinephrine intravenously (0.6+/-0.7 micro g/kg per minute) to achieve a mean arterial pressure of 77+/-12 mmHg. All patients received ventilatory support; two had acute oligoanuric renal failure treated by continuous hemodiafiltration. MEASUREMENTS AND RESULTS: We recorded heart rate, mean arterial pressure, central venous pressure, catecholamine dose, gas exchange, acid-base status, diuresis, C-reactive protein, white blood cell count, and Simplified Acute Physiological Score II. Except for transitory decreases in central venous pressure no changes were found in any variable. Six of seven patients died 5+/-3 days after the last plasmapheresis. CONCLUSIONS: In patients with norepinephrine-refractory septic shock no directional changes in measured variables were found in the first 24 h after a plasmapheresis. Thus, regarding the cost (ca. 1000 euros per separation) and possible risks the procedure needs rigorous evaluation even as "rescue therapy" in patients with septic shock.


Subject(s)
Cardiovascular System/drug effects , Norepinephrine/therapeutic use , Plasmapheresis , Shock, Septic/therapy , APACHE , Adult , Aged , Aged, 80 and over , Female , Hemodynamics/drug effects , Hospitals, Community , Humans , Male , Middle Aged , Norepinephrine/pharmacology , Plasmapheresis/adverse effects , Retrospective Studies , Shock, Septic/drug therapy , Shock, Septic/physiopathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...