Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.658
Filter
1.
Rinsho Ketsueki ; 65(6): 498-501, 2024.
Article in Japanese | MEDLINE | ID: mdl-38960647

ABSTRACT

A 43-year-old man with pancytopenia was diagnosed with acute promyelocytic leukemia (APL). On the first day of induction therapy with all-trans retinoic acid (ATRA) alone, he presented with high fever and was found to have coronavirus disease 2019 (COVID-19) infection by SARS-CoV2 antigen test. While it is generally recommended to delay treatment for APL patients with COVID-19 unless urgent APL treatment is required, this patient needed to continue treatment due to APL-induced disseminated intravascular coagulation (DIC). Considering the challenge of distinguishing between differentiation syndrome (DS) and COVID-19 exacerbation, the ATRA dosage was reduced to 50%. The patient was able to continue treatment without development of DS or exacerbation of DIC, leading to his recovery from COVID-19 and remission of APL.


Subject(s)
COVID-19 , Leukemia, Promyelocytic, Acute , Remission Induction , Tretinoin , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/complications , Tretinoin/administration & dosage , Tretinoin/therapeutic use , Male , Adult , COVID-19/complications , Treatment Outcome , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology
2.
Biochem Biophys Res Commun ; 724: 150140, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38852506

ABSTRACT

Sepsis is a severe inflammatory disease characterized by cytokine storm, often accompanied by disseminated intravascular coagulation (DIC). PANoptosis is a novel form of cell death triggered by cytokine storms, characterized by a cascade reaction of pyroptosis, apoptosis, and necroptosis. It exists in septic platelets and is closely associated with the onset and progression of DIC. However, there remains an unmet need for drugs targeting PANoptosis. The anti-PANoptosis effect of myricetin was predicted using network pharmacology and confirmed through molecular docking. In vitro platelet activation models demonstrated that myricetin significantly attenuated platelet particle release, integrin activation, adhesion, spreading, clot retraction, and aggregation. Moreover, in a sepsis model, myricetin reduced inflammatory infiltration in lung tissue and platelet activation while improving DIC. Additionally, whole blood sequencing samples from sepsis patients and healthy individuals were analyzed to elucidate the up-regulation of the PANoptosis targets. Our findings demonstrate the inhibitory effect of myricetin on septic platelet PANoptosis, indicating its potential as a novel anti-cellular PANoptosis candidate and therapeutic agent for septic DIC. Furthermore, our study establishes a foundation for utilizing network pharmacology in the discovery of new drugs to treat various diseases.


Subject(s)
Blood Platelets , Disseminated Intravascular Coagulation , Flavonoids , Sepsis , Flavonoids/pharmacology , Flavonoids/therapeutic use , Sepsis/drug therapy , Sepsis/blood , Humans , Blood Platelets/drug effects , Blood Platelets/metabolism , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Disseminated Intravascular Coagulation/pathology , Disseminated Intravascular Coagulation/blood , Animals , Male , Molecular Docking Simulation , Platelet Activation/drug effects , Mice, Inbred C57BL , Mice , Pyroptosis/drug effects
3.
ACS Nano ; 18(24): 15517-15528, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38836363

ABSTRACT

Disseminated intravascular coagulation (DIC) is a pathologic state that follows systemic injury and other diseases. Often a complication of sepsis or trauma, DIC causes coagulopathy associated with paradoxical thrombosis and hemorrhage. DIC upregulates the thrombotic pathways while simultaneously downregulating the fibrinolytic pathways that cause excessive fibrin deposition, microcirculatory thrombosis, multiorgan dysfunction, and consumptive coagulopathy with excessive bleeding. Given these opposing disease phenotypes, DIC management is challenging and includes treating the underlying disease and managing the coagulopathy. Currently, no therapies are approved for DIC. We have developed clot-targeted therapeutics that inhibit clot polymerization and activate clot fibrinolysis to manage DIC. We hypothesize that delivering both an anticoagulant and a fibrinolytic agent directly to clots will inhibit active clot polymerization while also breaking up pre-existing clots; therefore, reversing consumptive coagulopathy and restoring hemostatic balance. To test this hypothesis, we single- and dual-loaded fibrin-specific nanogels (FSNs) with antithrombinIII (ATIII) and/or tissue plasminogen activator (tPA) and evaluated their clot preventing and clot lysing abilities in vitro and in a rodent model of DIC. In vivo, single-loaded ATIII-FSNs decreased fibrin deposits in DIC organs and reduced blood loss when DIC rodents were injured. We also observed that the addition of tPA in dual-loaded ATIII-tPA-FSNs intensified the antithrombotic and fibrinolytic mechanisms, which proved advantageous for clot lysis and restoring platelet counts. However, the addition of tPA may have hindered wound healing capabilities when an injury was introduced. Our data supports the benefits of delivering both anticoagulants and fibrinolytic agents directly to clots to reduce the fibrin load and restore hemostatic balance in DIC.


Subject(s)
Disseminated Intravascular Coagulation , Tissue Plasminogen Activator , Tissue Plasminogen Activator/pharmacology , Tissue Plasminogen Activator/administration & dosage , Tissue Plasminogen Activator/chemistry , Animals , Disseminated Intravascular Coagulation/drug therapy , Nanogels/chemistry , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/administration & dosage , Humans , Rats , Fibrin/metabolism , Fibrin/chemistry , Antithrombins/pharmacology , Antithrombins/chemistry , Antithrombins/administration & dosage , Mice , Male , Thrombosis/drug therapy , Drug Delivery Systems , Blood Coagulation/drug effects
4.
Intensive Care Med ; 50(6): 849-860, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38748265

ABSTRACT

Hematological malignancies may require rapid-onset treatment because of their short doubling time, notably observed in acute leukemias and specific high-grade lymphomas. Furthermore, in targeted onco-hematological scenarios, chemotherapy is deemed necessary as an emergency measure when facing short-term, life-threatening complications associated with highly chemosensitive hematological malignancies. The risks inherent in the disease itself, or in the initiation of treatment, may then require admission to the intensive care unit (ICU) to optimize monitoring and initial management protocols. Hyperleukocytosis and leukostasis in acute leukemias, tumor lysis syndrome, and disseminated intravascular coagulation are the most frequent onco-hematological complications requiring the implementation of emergency chemotherapy in the ICU. Chemotherapy must also be started urgently in secondary hemophagocytic lymphohistiocytosis. Tumor-induced microangiopathic hemolytic anemia and plasma hyperviscosity due to malignant monoclonal gammopathy represent infrequent yet substantial indications for emergency chemotherapy. In all cases, the administration of emergency chemotherapy in the ICU requires close collaboration between intensivists and hematology specialists. In this review, we provide valuable insights that aid in the identification and treatment of patients requiring emergency chemotherapy in the ICU, offering diagnostic tools and guidance for their overall initial management.


Subject(s)
Hematologic Neoplasms , Intensive Care Units , Humans , Intensive Care Units/organization & administration , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/complications , Tumor Lysis Syndrome/etiology , Tumor Lysis Syndrome/drug therapy , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/adverse effects , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Lymphohistiocytosis, Hemophagocytic/drug therapy , Lymphohistiocytosis, Hemophagocytic/complications
5.
Ann Palliat Med ; 13(3): 477-495, 2024 May.
Article in English | MEDLINE | ID: mdl-38735696

ABSTRACT

BACKGROUND: Antithrombin is a small plasma glycoprotein synthesized in the liver that belongs to the serpin family of serine protease inhibitors and inactivates several enzymes in the coagulation pathway. It plays a leading major factor on coagulation pathway, therefore administration of antithrombin is essential to treat serious clinical conditions such as disseminated intravascular coagulation (DIC). Despite the theoretical benefits of antithrombin supplementation, the optimal antithrombin activity for heparin efficacy and the benefits of antithrombin supplementation in various disease entities are not yet fully understood. METHODS: The strict administration guidelines on antithrombin III in cases of DIC by the National Health Insurance Service and the Ministry of Food and Drug Safety complied as follows: antithrombin levels below 20 mg/dL in adults; antithrombin activity below 70% of normal in adults; total administration period of antithrombin must be carefully limited to within maximum 3 days, and the total administration dose must be below 7,000 international unit (IU), (loading dose, 1,000 IU in 1 hour: maintenance dose, 500 IU every 6 hours for 3 days). RESULTS: We identified 76 eligible for analysis according to the above-mentioned criteria in our institution (male/female, 59/17). Forty-four were identified to the non-survivor group and 32 patients were recognized as the survivor group. The baseline parameters in the non-survivor and survivor groups were comparable with no significant differences in age (66.5±18.1 vs. 66.0±16.2 years, P=0.90), sex (32/12 vs. 27/5, P=0.35), hospital length of stay (31.1±34.5 vs. 31.2±26.1 days, P=0.99), sequential organ failure assessment (SOFA) (7.3±2.5 vs. 6.6±2.0, P=0.22), simplified acute physiology score II (SAPS II) (46.0±8.8 vs. 43.5±9.2, P=0.23), cause for DIC (P=0.95), and underlying disease (P=0.38). The levels of antithrombin III on the day just before the administration significantly lower in the non-survivor groups than in the survivor groups (50.1%±13.6% vs. 57.6%±12.5%, P=0.01). The hemoglobin level in the 2nd day and 7th day after antithrombin III administration was significantly different between the non-survivor and survivor groups (9.9±1.9 vs. 11.0±2.0 g/dL, P=0.01, and 9.4±1.8 vs. 10.5±1.6 g/dL, P=0.006). The antithrombin III levels on the day of administration [area under the curve (AUC) =0.672] demonstrated significantly better prediction of mortality than the A antithrombin III levels on 1st day (AUC =0.552), the 2nd day (AUC =0.624), and 7th day (AUC =0.593). CONCLUSIONS: Our study suggests that the antithrombin administration may be effective tools for DIC treatment, and may be more positively considered, especially in the cases of DIC, which is a frequent complication of septic shock, sepsis, and other critical disease entities and which is associated with a high level of mortality. Furthermore, our study also suggests that the total doses and periods of antithrombin administration, which recommended by national guidelines, may be insufficient, therefore prolongation of period and increase of total dose of antithrombin supplement might be necessary.


Subject(s)
Antithrombin III , Disseminated Intravascular Coagulation , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Disseminated Intravascular Coagulation/drug therapy , Longitudinal Studies , Retrospective Studies , Treatment Outcome
6.
Clin Nutr ; 43(6): 1372-1383, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38678822

ABSTRACT

BACKGROUND & AIMS: Sepsis-induced disseminated intravascular coagulation (DIC) is characterised by abnormal blood clotting resulting from severe infection, contributing to organ dysfunction in sepsis. Resolvin D1 (RvD1) is an endogenous lipid mediator, synthesised from the omega-3 polyunsaturated fatty acid (PUFA) docosahexaenoic acid (DHA) through enzymatic processes involving 15-LOX and 5-LOX. RvD1 is recognised for its protective properties against various inflammatory conditions. This study aims to investigate its potential to modulate coagulation dysfunction in sepsis and to evaluate coagulation disorders in septic patients. METHODS: Sepsis models were established by intraperitoneal injection LPS (20 mg/kg) or cecal ligation and puncture (CLP) followed by injection of RvD1 (10 µg/kg) or saline. The impact of RvD1 on coagulation dysfunction was assessed by clotting time and coagulation indicators such as TAT, D-dimer, PAI-1, and fibrinogen. The activity of the coagulation system in vivo was observed by evaluating dynamic microcirculation, platelets and thrombin in mice using intravital microscopy. The effect of RvD1 on pyroptosis was investigated by measuring NOD-like receptor protein 3 (NLRP3), Caspase-1, Caspase-11, and Gasdermin D (GSDMD) levels via western blot. Caspase-1 knockout mice, GSDMD knockout mice and bone marrow-derived macrophages (BMDMs) were used to elucidate the underlying mechanisms. Lastly, the concentration of RvD1 in plasma from septic patients was quantified to explore its relationship with coagulation and pyroptosis. RESULTS: RvD1 significantly attenuated coagulation dysfunction in septic mice induced by LPS and CLP, and inhibited Caspase-1/GSDMD-dependent pyroptosis in septic mice and bone marrow-derived macrophages. In septic patients, the plasma concentrations of RvD1 was negatively correlated with both coagulation-related indicators and markers of GSDMD activation. CONCLUSION: The results suggest that RvD1 can improve coagulation dysfunction in sepsis by regulating the Caspase-1/GSDMD pyroptotic pathway. Additionally, the concentration of RvD1 in septic patient plasma is related to prognosis and DIC development. RvD1 could be a potential biomarker and a promising therapeutic alternative in sepsis-induced DIC.


Subject(s)
Caspase 1 , Disseminated Intravascular Coagulation , Docosahexaenoic Acids , Mice, Inbred C57BL , Phosphate-Binding Proteins , Pyroptosis , Sepsis , Animals , Sepsis/complications , Sepsis/drug therapy , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Mice , Male , Caspase 1/metabolism , Humans , Docosahexaenoic Acids/pharmacology , Pyroptosis/drug effects , Phosphate-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Disease Models, Animal , Female , Middle Aged , Mice, Knockout , Signal Transduction/drug effects , Aged , Gasdermins
7.
Haemophilia ; 30(3): 836-844, 2024 May.
Article in English | MEDLINE | ID: mdl-38523253

ABSTRACT

BACKGROUND: Emicizumab (Emi) is used as haemostatic prophylaxis for patients with haemophilia A (PwHA). Disseminated intravascular coagulation (DIC) is a condition characterized by persistent systemic activation of coagulation, but there is yet no information on coagulation and fibrinolysis potentials in Emi-treated PwHA with DIC. AIM: To examine the effect of Emi on coagulation and fibrinolysis potentials in HA-model DIC plasmas. METHODS: Plasma from a patient with sepsis-DIC (seven patients) was treated with anti-factor (F)VIII monoclonal antibody (HA-model DIC plasma) and incubated with Emi (50 µg/mL). The plasma was then assessed using clot-fibrinolysis waveform analysis (CFWA). Coagulation and fibrinolysis parameters were expressed as ratios relative to normal plasma (|min1|-ratio and |FL-min1|-ratio, respectively). PATIENTS AND RESULTS: In case 1, coagulant potential was slightly high and fibrinolytic potential was extremely low, presenting a coagulant-dominant state (|min1|-ratio/|FL-min1|-ratio: 1.1/.38). In cases 2-5, fibrinolytic potential was not suppressed, but there were marked hypercoagulant potentials, indicating relative coagulant-dominant states. In case 6, coagulant and fibrinolytic potentials were increased but well balanced (|min1|-ratio/|FL-min1|-ratio: 1.38/1.28). In case 7, both potentials were severely deteriorated in not only CFWA but also the thrombin/plasmin generation assay. The addition of Emi into the HA-model DIC plasmas increased |min1|-ratio values in all cases, but the coagulant potentials did not exceed the initial ones (DIC plasma before treatment with anti-FVIII antibody). CONCLUSIONS: The presence of Emi in the HA-model DIC plasma improved coagulation potentials, but did not increase coagulation potentials beyond those of DIC plasma in non-HA states.


Subject(s)
Antibodies, Bispecific , Antibodies, Monoclonal, Humanized , Blood Coagulation , Disseminated Intravascular Coagulation , Fibrinolysis , Humans , Fibrinolysis/drug effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Disseminated Intravascular Coagulation/blood , Antibodies, Bispecific/therapeutic use , Antibodies, Bispecific/pharmacology , Blood Coagulation/drug effects , Male , Middle Aged , Factor VIII/therapeutic use , Factor VIII/pharmacology , Factor VIII/immunology , Aged , Female , Adult
8.
BMC Cardiovasc Disord ; 24(1): 78, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38287252

ABSTRACT

BACKGROUND: Management of the enhanced-fibrinolytic type of disseminated intravascular coagulation (DIC) caused by aortic disorders is the two strategies of surgical intervention and medical treatment based on the patient's age and comorbidities. CASE PRESENTATION: An 81-year-old woman with a history of two previous aortic surgeries and chronic heart and renal failure was admitted for uncontrollable subcutaneous hemorrhage. The hemorrhage was caused by the enhanced-fibrinolytic type of disseminated intravascular coagulation (DIC) caused by periprosthetic graft hematoma after aortic replacement for Stanford type A aortic dissection. Open thoracic hemostasis temporarily controlled the subcutaneous hemorrhage, but she was readmitted for the recurrence seven months after discharge. On the second admission, the combination of anticoagulant and antifibrinolytic agents was successful. CONCLUSION: Management of the enhanced-fibrinolytic type of DIC caused by aortic disorders is important of a successful combination of surgical and medical therapy tailored the patient's condition.


Subject(s)
Aortic Aneurysm , Aortic Dissection , Disseminated Intravascular Coagulation , Renal Insufficiency , Female , Humans , Aged, 80 and over , Aortic Aneurysm/complications , Disseminated Intravascular Coagulation/diagnosis , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Aortic Dissection/complications , Aortic Dissection/diagnostic imaging , Aortic Dissection/surgery , Hemorrhage , Renal Insufficiency/complications
10.
Int J Hematol ; 119(4): 416-425, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38270783

ABSTRACT

BACKGROUND: Recombinant human soluble thrombomodulin (rhTM) is commonly used in Japan to treat disseminated intravascular coagulation (DIC), but its efficacy compared with other anticoagulants is unclear. We conducted a systematic review and meta-analysis to investigate this issue in DIC patients with hematological malignancies. METHODS: We searched PubMed, Cochrane, and Scopus for prospective and retrospective studies evaluating the efficacy and safety of rhTM in DIC patients with hematological malignancies between April 2008 and April 2023. We performed a systematic review and meta-analysis evaluating recovery from DIC, hemorrhagic adverse events (AEs), and overall survival (OS). RESULTS: We analyzed one prospective (64 patients) and seven retrospective studies (209 patients). Use of rhTM was associated with a higher rate of recovery from DIC (OR: 2.25 [1.09-4.63] and 1.98 [1.12-3.50] in prospective and retrospective studies, respectively; same order below) and fewer hemorrhagic AEs (OR: 0.83 [0.30-2.30] and 0.21 [0.08-0.57]). rhTM did not improve OS (OR: 1.06 [0.42-2.66] and 1.72 [0.87-3.39]), although the incidence of hemorrhagic death was lower in the rhTM group (0 of 94 patients). CONCLUSION: Use of rhTM in patients with hematological malignancy-associated DIC is strongly expected to be effective and safe.


Subject(s)
Disseminated Intravascular Coagulation , Hematologic Neoplasms , Sepsis , Humans , Retrospective Studies , Prospective Studies , Thrombomodulin/therapeutic use , Treatment Outcome , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Sepsis/complications , Recombinant Proteins/adverse effects , Hematologic Neoplasms/complications , Hematologic Neoplasms/drug therapy , Hemorrhage
12.
Trials ; 25(1): 4, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38167115

ABSTRACT

BACKGROUND: Disseminated intravascular coagulation (DIC) occurs in 30-50% of septic patients and contributes to high mortality in the intensive care unit (ICU). However, there are few proven interventions for coagulation disorder management in sepsis. Experimental and clinical data have demonstrated that sepsis could benefit from unfractionated heparin (UFH) treatment. To date, there are no large multicenter trials to determine the safety and efficacy of UFH in septic patients with suspected DIC. METHODS: A multicenter, double-blinded, placebo-controlled randomized trial is designed to recruit 600 patients who met sepsis 3.0 criteria and suspected DIC. Participants will be randomized (1:1) to receive UFH or saline via continuous intravenous administration for 7 days within 6 h of enrolment. The primary outcome is ICU mortality. The secondary outcome includes 28-day all-cause mortality, the improvement of Sequential Organ Failure Assessment scores, and the incidence of major hemorrhage. Investigators, participants, and statisticians will be blinded to the allocation. DISCUSSION: The HepSIC trial is to evaluate the efficacy and safety of UFH on sepsis-related DIC across different areas of China. The small dosage of UFH administration would offer a new potential approach for treating sepsis-related coagulation disorders. ETHICS AND DISSEMINATION: Ethical approval was granted by all the ethics committees of 20 participant centers. Results will be disseminated via peer-reviewed publications and presented at conferences. TRIAL REGISTRATION: ClinicalTrials.gov NCT02654561. Registered on 13 January 2016.


Subject(s)
Blood Coagulation Disorders , Disseminated Intravascular Coagulation , Sepsis , Humans , Heparin/adverse effects , Disseminated Intravascular Coagulation/diagnosis , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Sepsis/complications , Sepsis/diagnosis , Sepsis/drug therapy , Blood Coagulation Disorders/drug therapy , Hemorrhage/drug therapy , Randomized Controlled Trials as Topic , Multicenter Studies as Topic
13.
Toxicon ; 238: 107563, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38141969

ABSTRACT

This case report summarizes an envenomation by the Mangshan pit viper (Protobothrops mangshanensis), a rare, endangered, venomous snake endemic to Mount Mang of China, and the first reported use of Hemato Polyvalent antivenom (HPAV) for this species. The snakebite occurred in a United States zoo to a 46-year-old male zookeeper. He presented via emergency medical services to a tertiary center after sustaining a single P. mangshanensis bite to the abdomen and was transported with antivenom from the zoo. Within 2 hours of envenomation, he developed oozing of sanguineous fluid and ecchymosis at the puncture site, and about 4 hours post-bite, was treated with HPAV. His coagulation profile fluctuated with the following pertinent peak/nadir laboratory values and corresponding hospital day (HD): undetectable fibrinogen levels, d-dimer 8.89 mg/L and 7.43 mg/L, and INR 2.97 and 1.46 on HD zero and three, respectively. Other peak/nadir values included hemoglobin 9.7 g/dL and creatinine phosphokinase 2410 U/L on HD four and platelets 81 × 109/L on HD seven. The patient received a total of 30 vials of HPAV over 5 days and 1 unit of cryoprecipitate on HD six. Upon discharge on HD eight, laboratory studies were normalizing, except for platelets, and edema stabilized. This case describes an acute, recurrent, and prolonged venom-induced consumptive coagulopathy despite prompt administration and repeated doses of HPAV.


Subject(s)
Crotalid Venoms , Crotalinae , Disseminated Intravascular Coagulation , Snake Bites , Male , Animals , Humans , Middle Aged , Antivenins/therapeutic use , Snake Bites/drug therapy , Blood Coagulation Tests , Disseminated Intravascular Coagulation/chemically induced , Disseminated Intravascular Coagulation/drug therapy , Crotalid Venoms/toxicity , Viper Venoms
14.
J Pak Med Assoc ; 73(12): 2476-2479, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38083937

ABSTRACT

Kasabach-Merritt syndrome is a rare condition, characterised by the presence of an enlarging vascular tumour associated with thrombocytopenia, microangiopathic haemolytic anaemia and consumptive coagulopathy. The syndrome manifests in infancy, with high morbidity and mortality rates. No standard guidelines have been established for the treatment of Kasabach-Merritt syndrome to date. To existing literature we add this report of a four-month-old female child with Kasabach-Merritt syndrome who was successfully treated with propranolol and vincristine. This drug combination helped reverse the severe thrombocytopenia as well as decrease in size of her haemangioma. Management of Kasabach-Merritt syndrome continues to be a challenge, with varying response to first line drugs. Early diagnosis and initiation of treatment in a closely monitored setting is essential to ensure good outcomes. Since this is a relatively rare condition and large studies are not feasible, documenting treatment experience for single cases or small series becomes even more important.


Subject(s)
Disseminated Intravascular Coagulation , Hemangioma , Kasabach-Merritt Syndrome , Female , Humans , Infant , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Hemangioma/complications , Hemangioma/drug therapy , Kasabach-Merritt Syndrome/complications , Kasabach-Merritt Syndrome/drug therapy , Kasabach-Merritt Syndrome/diagnosis , Propranolol/therapeutic use , Vincristine/therapeutic use
15.
Hinyokika Kiyo ; 69(11): 309-314, 2023 Nov.
Article in Japanese | MEDLINE | ID: mdl-38031329

ABSTRACT

Severe urinary tract infections occasionally cause sepsis and disseminated intravascular coagulation (DIC). We examined the efficacy of recombinant thrombomodulin (rTM) for treating DIC caused by urosepsis. We enrolled 40 patients who were diagnosed with DIC caused by urosepsis at our hospital between April 2018 and May 2022. Twenty-six patients were treated with rTM (rTM group), while 14 patients did not receive rTM (non-rTM group). The DIC score before treatment in the rTM group was significantly higher than that in the non-rTM group (P<0.01). There was no significant difference in disease-specific survival between the two groups. There was a significant improvement in DIC scores on days 1-3 after administering rTM. However, the duration of DIC in the rTM group was significantly longer than that in the non-rTM group (P=0.038). The administration of rTM may have benefits in patients with DIC caused by urosepsis.


Subject(s)
Disseminated Intravascular Coagulation , Sepsis , Thrombomodulin , Urinary Tract Infections , Humans , Disseminated Intravascular Coagulation/complications , Disseminated Intravascular Coagulation/drug therapy , Recombinant Proteins/therapeutic use , Sepsis/complications , Sepsis/drug therapy , Thrombomodulin/therapeutic use , Treatment Outcome , Urinary Tract Infections/complications , Urinary Tract Infections/drug therapy
16.
Shock ; 60(2): 221-226, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37342874

ABSTRACT

ABSTRACT: Thrombomodulin alfa (TM alfa) has been shown effective for treatment of disseminated intravascular coagulation (DIC) associated with sepsis, although the optimal therapeutic plasma concentration has not been clarified. In the present study, the plasma trough concentration of TM alfa in septic patients with DIC was determined, then the cutoff value for that concentration showing influence on treatment outcome was calculated using a receiver operating characteristic curve. With a cutoff value of 1,010, the area under the curve of the receiver operating characteristic was 0.669 (95% confidence interval, 0.530-0.808), with sensitivity of 0.458 and specificity of 0.882. To evaluate its accuracy, patients were divided into those above or below the cutoff value, and 90-day survival rates were compared. The above-cutoff group showed a significantly higher 90-day survival rate (91.7%) as compared with the below-cutoff group (63.4%) ( P = 0.017), with a hazard ratio of 0.199 (95% confidence interval, 0.045-0.871). Interestingly, the incidence of hemorrhagic adverse effects was not significantly different between the groups. Based on these results, the recommended plasma trough concentration of TM alfa for treatment of septic DIC is 1,010 ng/mL, which should minimize the risk of severe bleeding while maximizing the therapeutic effect.


Subject(s)
Disseminated Intravascular Coagulation , Sepsis , Humans , Thrombomodulin/therapeutic use , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Sepsis/complications , Sepsis/drug therapy , Anticoagulants/therapeutic use , Treatment Outcome
17.
Chin J Traumatol ; 26(5): 297-302, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37321903

ABSTRACT

The Sepsis Coagulopathy Asahi Recombinant LE Thrombomodulin (SCARLET) trial has many defects, and thus cannot be the terminator of recombinant thrombomodulin (rTM). On the contrary, it provides sufficient evidence for further research. Based on analysis focusing on the failure of SCARLET and several previous anticoagulant studies, it is most important for new studies to grasp the following two points: (1) The enrolled cases should have sufficient disease severity and a clear standard for disseminated intravascular coagulation; (2) Heparin should not be used in combination with the investigated drugs. Multiple post-hoc analyses show that no combination of heparin will not increase the risk of thromboembolism. In fact, the combination of heparin can mask the true efficacy of the investigated drug. Due to the complexity of sepsis treatment and the limitations of clinical studies, the results of all treatment studies should be repeatedly verified, rather than be determined at one stroke. Some research conclusions contrary to disease physiology, pharmacology and clinical practice may be deceptive, and should be cautious rather than be simply accepted. On the other hand, the dissenting voices in the "consensus" scene are often well discussed by the authors and should be highly valued.


Subject(s)
Blood Coagulation Disorders , Disseminated Intravascular Coagulation , Sepsis , Humans , Anticoagulants/therapeutic use , Thrombomodulin/therapeutic use , Disseminated Intravascular Coagulation/drug therapy , Sepsis/drug therapy , Heparin/therapeutic use , Recombinant Proteins
18.
Thromb Res ; 226: 165-172, 2023 06.
Article in English | MEDLINE | ID: mdl-37182388

ABSTRACT

BACKGROUND: Recombinant human soluble thrombomodulin (rhTM) is used to treat sepsis-induced disseminated intravascular coagulation (DIC). However, no consistent clinical guidelines exist regarding the administration of rhTM in patients with sepsis-induced DIC. Therefore, we conducted this meta-analysis to evaluate the efficacy and safety of rhTM therapy in patients with sepsis-induced DIC. METHODS: EMBASE, PubMed, Scopus, Ichushi, and CINAHL databases were used to search for relevant articles that met the inclusion criteria of patients with sepsis-induced DIC treated with and without rhTM through November 2022. Mortality, DIC resolution, and incidence of bleeding complications were evaluated. DIC resolution was defined as the recovery from DIC after the start of DIC treatment. RESULTS: Of the 1697 citations identified for screening, 17 studies involving 2296 patients were included. Administering rhTM significantly reduced mortality (odds ratio (OR) 0.54, 95 % confidence interval (CI) 0.42-0.71) and improved DIC resolution (OR 2.88, 95 % CI 1.83-4.52). There were no significant differences in the incidence of bleeding complications between the rhTM and control groups (OR 0.92, 95 % CI 0.66-1.28). CONCLUSIONS: Our meta-analysis revealed that rhTM could reduce mortality and improve DIC resolution without increasing the risk of bleeding in patients with sepsis-induced DIC. Our findings suggest that rhTM is a relatively effective and safe anticoagulant for the treatment of sepsis-induced DIC. SUMMARY: Recombinant human soluble thrombomodulin reduced mortality without increasing the bleeding risk in the treatment of sepsis-induced disseminated intravascular coagulation.


Subject(s)
Disseminated Intravascular Coagulation , Sepsis , Humans , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Recombinant Proteins , Retrospective Studies , Sepsis/complications , Sepsis/drug therapy , Sepsis/diagnosis , Thrombomodulin/therapeutic use , Treatment Outcome
19.
Int J Clin Pharmacol Ther ; 61(7): 297-305, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37114512

ABSTRACT

OBJECTIVE: Recombinant human soluble thrombomodulin (rhsTM) is a therapeutic agent for sepsis-induced disseminated intravascular coagulation (DIC) and is associated with bleeding events. rhsTM is a renal excretion drug; however, information on the role of rhsTM in renal function is limited. MATERIALS AND METHODS: In this retrospective observational study, we assessed rhsTM-associated bleeding events according to the renal function of patients with sepsis-induced DIC. We analyzed the data of 79 patients administered a standard-dose of rhsTM for sepsis-induced DIC, at a single center. Patients were classified based on estimated glomerular filtration rate (eGFR). We measured fresh bleeding events following rhsTM administration, DIC score efficacy, and 28-day mortality. RESULTS: Fresh bleeding events were observed in 15 patients, with a significant difference in the eGFR, platelet count, and DIC scores. Furthermore, fresh bleeding events tended to increase with the deterioration of renal function (p = 0.039). The DIC scores in all renal function groups decreased after -rhsTM administration. Additionally, the 28-day mortality was less than 30% in all groups. CONCLUSION: Our results indicate that the effectiveness of the standard-dose of rhsTM is not related to renal function. However, standard-dose rhsTM therapy could potentially increase the risk of adverse bleeding events with severe renal function equivalent to G5.


Subject(s)
Disseminated Intravascular Coagulation , Sepsis , Humans , Disseminated Intravascular Coagulation/diagnosis , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Thrombomodulin/therapeutic use , Recombinant Proteins/adverse effects , Hemorrhage , Sepsis/complications , Sepsis/drug therapy , Kidney/physiology , Treatment Outcome
20.
In Vivo ; 37(3): 1389-1393, 2023.
Article in English | MEDLINE | ID: mdl-37103098

ABSTRACT

BACKGROUND/AIM: Bone marrow metastasis (BMM) of gastric cancer (GC) is complicated by disseminated intravascular coagulation syndrome (DIC), which is more prominent in poorly differentiated carcinoma. This is one of the first case reports of a slowly progressing BMM of GC after approximately 1 year of follow-up without treatment. CASE REPORT: A 72-year-old woman underwent total gastrectomy and splenectomy for GC in February 2012. The pathological diagnosis was that of a moderately differentiated adenocarcinoma. Five years later in December 2017, she developed anemia; however, its cause remained unknown. Due to worsening of the anemia, the patient visited the Kakogawa Central City Hospital in October 2018. Bone marrow biopsy revealed an infiltration of caudal type homeobox 2-positive cancer cells, and our diagnosis was BMM of GC. There was no DIC. The incidence of BMM is high in well- or moderately differentiated breast cancer but rarely causes DIC. CONCLUSION: As with breast cancer, in moderately differentiated cancer cells, BMM of GC may progress slowly after the appearance of symptoms without causing DIC.


Subject(s)
Bone Marrow Neoplasms , Bone Neoplasms , Breast Neoplasms , Disseminated Intravascular Coagulation , Stomach Neoplasms , Female , Humans , Aged , Bone Marrow/pathology , Stomach Neoplasms/pathology , Bone Marrow Neoplasms/drug therapy , Bone Marrow Neoplasms/secondary , Disseminated Intravascular Coagulation/complications , Disseminated Intravascular Coagulation/drug therapy , Bone Neoplasms/secondary , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...