Pacritinib, a Dual JAK2/FLT3 Inhibitor: An Integrated Efficacy and Safety Analysis Of Phase II Trial Data In Patients With Primary and Secondary Myelofibrosis (MF) and Platelet Counts ≤100,000/µl

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 395-395 ◽  
Author(s):  
Srdan Verstovsek ◽  
James P. Dean ◽  
Paul Cernohous ◽  
Rami S Komrokji ◽  
John F. Seymour ◽  
...  

Abstract Background and Aims A platelet count ≤100,000/µL is regarded as an independent prognostic marker for the development of leukemia and is associated with inferior survival in myelofibrosis (MF) (Huang et al. 2008). The approved JAK2 inhibitor, ruxolitinib, is associated with treatment-emergent thrombocytopenia, and patients with platelets ≤100,000/µL were excluded from the phase III COMFORT trials. Efficacy and safety data are limited in patients with MF and platelets ≤100,000/µL treated with ruxolitinib. Pacritinib (SB1518), an oral JAK2/FLT3 inhibitor, did not appear to be associated with clinically significant treatment emergent anemia or thrombocytopenia during early phase studies in patients with myeloid and lymphoid malignancies. This integrated analysis was performed to further evaluate the safety and efficacy of pacritinib in MF patients with baseline platelet counts of ≤100,000/µL. Methods The databases from 2 phase I/II clinical trials of pacritinib in advanced myeloid malignancies and MF were integrated to evaluate the rate of adverse events (AEs) and favorable spleen responses in patients with MF and thrombocytopenia. Results One hundred twenty-nine patients with advanced myeloid malignancies and MF were treated with pacritinib in phase I and II clinical trials. Sixty-five patients with MF were treated with 400 mg orally once daily in 2 phase II studies. Of these, 28 patients with MF [21 primary MF (75%), 5 post-polycythemia vera-MF, 2 post-essential thrombocytosis-MF] had baseline platelet counts of ≤100,000/µL (median: 58,500/µL, range: 15,000/µL – 97,000/µL). The median age was 69 years. Twenty three (82%) patients had the JAK2V617F mutation. During the phase 2 studies, spleen volume responses were assessed by MRI and are shown for all patients (Figure 1) and for patients with platelets ≤100,000/µL (Figure 2). Thirty-seven percent and 43% of all evaluable patients and patients with ≤100,000/µL platelets, respectively, had ≥35% reduction in spleen volume from baseline (Table). Among the 21 informative patients with allelic JAK2V617F quantification at more than 1 study visit, 3, including 2 patients with homozygous mutations, had ≥35% reduction in spleen volume and substantial reductions in allelic burden from baseline (mean reduction 64%, range 44 to 88%). In the 11 (17%) patients with MF and baseline platelet counts ≤50,000/μL, overall spleen volume reduction was 38% (3/8) in evaluable patients and the median decline in platelet count observed at study end was 6,000/µL. Integrated safety analysis for all 129 patients with advanced myeloid malignancies showed the most common adverse events (AEs) were gastrointestinal (GI), predominantly diarrhea, and most of these were grade 1 (43%) and grade 2 (26%). The time to onset of diarrhea was ≤30 days in the majority of those affected, but only 2% had drug discontinuation as a consequence. Among the 129 patients in the phase 1 and 2 trials, 57 patients had ≤100,000 platelets; of these 26 (46%) had no CTC grade change in hemoglobin (Hb) and platelet counts from baseline values. Hb and platelet counts improved by one grade in 16% and 18% of patients, respectively. One grade worsening was seen in 28% and 30% in Hb and platelet counts, respectively. Conclusions Pacritinib was well-tolerated in patients with MF, regardless of their baseline platelet counts. Grade 1 or 2 GI toxicities, particularly diarrhea, were the most common AEs. Spleen responses were similar in patients regardless of baseline platelet counts (≥100,000/µL vs. ≤100,000/µL). Notably, even patients with initial platelet counts ≤50,000/µL tolerated therapy and the majority maintained stable hemoglobin and platelet counts. A phase III trial will evaluate pacritinib versus best available therapies in patients with MF and platelet counts ≤100,000/µL. Disclosures: Dean: Cell Therapeutics, Inc: Employment. Cernohous:Cell Therapeutics, Inc: Employment. Mesa:Incyte, CTI, Gilead, Celgene, Genetech, NS Pharma, Lilly: Research Funding. Campbell:Cell Therapeutics, Inc: Employment. Caldwell:Cell Therapeutics, Inc: Employment. Wang:Cell Therapeutics, Inc.: Employment. Myint:Cell Therapeutics, Inc: Employment.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1622-1622
Author(s):  
Shijia Zhang ◽  
Yucai Wang ◽  
Larysa Sanchez ◽  
Narjust Duma ◽  
Victor Chang

Abstract Background: Ruxolitinib, a potent JAK1/JAK2 inhibitor, was approved by the FDA for the treatment of myelofibrosis (MF) and polycythemia vera (PV) in 2011 and 2014, respectively. The aim of this study was to evaluate the efficacy and safety data of single-agent ruxolitinib in patients with MF or PV through meta-analysis of reported clinical trials. Methods: PubMed and Web of Science databases were searched for clinical trials of ruxolitinib for the treatment of MF or PV through June 30th, 2015. Only clinical trials in which ruxolitinib was used as single-agent therapy for MF or PV were included in this meta-analysis. Pooled odds ratios (ORs) for proportion of patients with >= 35% reduction in spleen volume (primary outcome) and risk ratios (RRs) for dichotomous data with 95% confidence interval (CI) were analyzed using OpenMeta[Analyst] with a random effect model. Pooled ratios of adverse events of ruxolitinib therapy were also calculated. Results: Six clinical trials including 3 randomized controlled trials (RCTs) of 987 patients were identified and included in the analysis. Four studies were conducted in MF patients, and 2 studies in PV patients. Pooled data from the 3 RCTs (2 for MF and 1 for PV) showed that ruxolitinib, compared to placebo or standard therapy/best available therapy (BAT), significantly increased the proportion of patients with a reduction in spleen volume of 35% or more (OR = 79.55, 95% CI = 22.51-281.11, p < 0.001). Ruxolitinib improved the symptoms associated with MF or PV. Only the 2 RCTs for MF analyzed overall survival (OS), one of which showed OS benefit with ruxolitinib. Pooled data from all 6 trials showed that the most common hematologic adverse events were all grades anemia (71.8%) and thrombocytopenia (54.1%), regardless of whether or not they were deemed related to ruxolitinib therapy. Incidence of grades 3/4 anemia and thrombocytopenia was 30.2% and 15.3%, respectively, but rarely led to discontinuation of the drug. The most common non-hematologic adverse events were diarrhea (all grades = 18.1%, grades 3/4 = 1.1%), fatigue (all grades = 14.0%, grades 3/4 = 2.0%), peripheral edema (all grades = 12.0%, grades 3/4 = 0%), and dyspnea (all grades = 10.0%, grades 3/4 = 0.9%). Further analysis was conducted to evaluate the attributable risks of the adverse events using data from the 3 RCTs. Compared to placebo or standard therapy/BAT, ruxolitinib significantly increased the risk of developing all grades anemia (RR = 1.10, 95% CI = 1.03-1.17, p = 0.005), grades 3/4 anemia (RR = 1.87, 95% CI = 1.20-2.93, p = 0.006), all grades thrombocytopenia (RR = 2.04, 95% CI = 1.47-2.83, p < 0.001), but not grades 3/4 thrombocytopenia (RR = 2.04, 95% CI = 0.72-7.98, p = 0.153), all grades diarrhea (RR = 1.45, 95% CI = 0.96-2.20, p = 0.079), fatigue (RR = 0.865, 95% CI = 0.62-1.20, p = 0.382), peripheral edema (RR = 0.85, 95% CI = 0.61-1.18, p = 0.337), and dyspnea (RR = 1.25, 95% CI = 0.63-2.49, p = 0.520). Conclusions: Single-agent ruxolitinib therapy results in improvement of symptoms in MF or PV and significant reduction in spleen size compared to placebo or standard therapy/BAT. Anemia and thrombocytopenia were the most common hematologic adverse events, but rarely led to the discontinuation of therapy. Diarrhea, fatigue, peripheral edema and dyspnea were the most common non-hematologic adverse events, but not significant compared to placebo or standard therapy/BAT. Disclosures Chang: Johnson & Johnson: Other: Stock.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 6576-6576
Author(s):  
T. L. Koeneke ◽  
J. O. Armitage ◽  
P. J. Bierman ◽  
R. Bociek ◽  
J. M. Vose ◽  
...  

6576 Background: Arguments have been made against early phase clinical trials (CTs) as possibly being unethical because its risk may outweigh its potential benefits. Whether this is true in the light of newer biological treatment for cancer is unknown. We therefore examined the association between the incidence of serious adverse events according to type and sponsorship of CTs in pts with lymphoma. Methods: All IRB approved CTs at the University of Nebraska Medical Center from Jan 2000-June 2005 classified as therapeutic for lymphoma involving a biological agent were included. CTs were classified in two ways: by type of CTs (phase I vs II vs III) and sponsorship (Investigator-initiated vs Industry-initiated. Multivariate logistic regression was used to evaluate the association between types/sponsorship of CTs with the incidence of IRB serious adverse events (SAE; no vs yes) and fatal adverse events (FAE; no vs yes) while adjusting for age, sex, race, lymphoma type and stage, interval from dx to tx, co-morbid conditions, and previous tx. Results: 357 pts with lymphoma enrolled in 29 CTs were included. The median age of pt was 54y (21–88). 41% of the pts had follicular lymphoma, 36% diffuse large cell, 14% mantle cell and 9% were other types. 59% had Stage IV lymphoma. 71% of the pts participated in investigator-initiated CTs, while 29% participated in industry-initiated CTs. 21% of pts were enrolled in phase I, 65% in phase II and 14% in phase III studies. SAEs were seen in 49 pts (14%), while FAEs occurred in 13 pts (4%). Multivariate analysis showed the risk of having SAE was independent of the type or sponsor of CTs. Additionally, the risk of FAEs was not associated with the type of CTs. However, the risk of having FAEs was less in investigator- iniatiated CTs than in industry-iniatiated trials (Odds Ratio: 0.13 (95% CI, 0.03–0.61, p = 0.01). Conclusions: Our study showed that in CTs involving biological treatments, the incidence of SAEs was not associated with the type or sponsor of CTs suggesting that use of biological agents in phase I studies may have similar risks to phase II/III trials. Further studies should be done in other types of malignancies to evaluate further the decrease frequency of FAEs seen in investigator-initiated trials. No significant financial relationships to disclose.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. e20533-e20533
Author(s):  
D. E. Tsai ◽  
W. Wang ◽  
R. Reshef ◽  
D. Vogl ◽  
E. Stadtmauer ◽  
...  

e20533 Background: Bexarotene (Bex) is an oral retinoid X receptor agonist with activity against cutaneous T cell lymphoma and currently under investigation for other malignancies. In patients receiving this agent for acute myeloid leukemia (AML), we noted increases in platelet counts. We therefore reviewed the available clinical trial data on Bex and its effects on platelet counts. Methods: We analyzed platelet count data from 3 Bex clinical trials encompassing non-small cell lung cancer (NSCLC) and AML. Results: In two phase III trials of Bex in NSCLC, patients underwent carboplatin + paclitaxel (CarP, n=587) or cisplatin + vinorelbine (CisV, n=613) and were randomized to receive concurrent Bex or placebo. More patients on Bex than on placebo had an increase in platelet count of at least 50 K/uL (55% vs. 27% for CarP, p<0.0001; 81% vs. 66% for CisV, p<0.0001) over pre-treatment baseline. The median increase in platelet count was higher on Bex than on placebo (69 vs 0 K/uL for CarP, p<0.0001; 168 vs. 95 K/uL for CisV, p<0.0001) and was maintained while on treatment. In both NSCLC trials, the median time to platelet increase >50 K/uL on Bex was 22 days. Similar findings were seen in a phase I monotherapy trial in AML where 5/18 (28%) patients achieved platelet transfusion independence with peak platelet counts of 40–91 K/uL. Conclusions: Clinically significant increases in platelet counts were seen in all 3 clinical trials examined. These data suggest that Bex improves platelet counts in patients with a variety of cancer types, both as monotherapy and with concurrent chemotherapy. Its effect on megakaryopoiesis and its potential role as a supportive care measure should be further evaluated. [Table: see text]


2021 ◽  
Vol 10 ◽  
Author(s):  
Li Sun ◽  
Yi-Jia Guo ◽  
Jun Song ◽  
Yan-Ru Wang ◽  
Shu-Ling Zhang ◽  
...  

PurposeThe role of neoadjuvant epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI) targeted therapy for patients with EGFR-mutant non-small cell lung cancer (NSCLC) has not been clarified. A pooled analysis of prospective clinical trials was conducted to evaluate the efficacy and safety of neoadjuvant EGFR-TKI therapy.MethodsThe PubMed, Embase, Web of Science, and Cochrane Library databases, as well as meeting abstracts were searched for prospective clinical trials evaluating the efficacy and safety of neoadjuvant EGFR-TKI for treatment of EGFR-mutant NSCLC. The main outcomes included the objective response rate (ORR), downstaging rate, surgical resection rate (SRR), pathologic complete response (pCR) rate, progression-free survival (PFS), and adverse events.ResultsA total of five, phase II, prospective, clinical trials involving 124 patients with resectable or potentially resectable EGFR-mutant NSCLC treated with neoadjuvant erlotinib or gefitinib treatment were included in this pooled analysis. The median neoadjuvant medication time was 42 (range, 21–56) days and the median time of response evaluation was 45 (range, 42–56) days. The pooled ORR was 58.5% [95% confidence interval (CI), 45.5%–71.8%] and the surgical resection and complete resection (R0) rates were 79.9% (95% CI, 65.3%–94.5%) and 64.3% (95% CI, 43.8%–84.8%), respectively. In the stage IIIA subgroup (n = 68), the pooled ORR, SRR, and R0 rate were 51.4%, 72.9%, and 57.0%, respectively, while the downstaging and pCR rates were 14.0% and 0.0%, respectively. The pooled median PFS and overall survival were 13.2 and 41.9 months, respectively. Of the most common grade 3/4 adverse events in the overall group, the incidences of hepatotoxicity and skin rash were 5.3% and 14.7%, respectively. The most commonly reported postoperative complications were lung infection, arrhythmia, and pneumothorax.ConclusionNeoadjuvant EGFR-TKI therapy provides a feasible treatment modality for patients with resectable or potentially resectable EGFR-mutant NSCLC, with satisfactory surgical outcomes and low toxicity. Although further phase III clinical trials are needed to confirm these findings, it is necessary to explore the feasibility of a more effective EGFR-TKI combination neoadjuvant therapy given the modest downgrade and pCR rates for EGFR-TKI alone.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3285-3285
Author(s):  
Nongnuch Sirachainan ◽  
Usanarat Anurathapan ◽  
Ampaiwan Chuansumrit ◽  
Duantida Songdej ◽  
Pakawan Wongwerawattanakoon ◽  
...  

Abstract Abstract 3285 Introduction: Chronic ITP accounts for 20–30% of children with ITP. Treatment includes immunosuppressive agents and splenectomy to maintain platelet count above 20,000/uL. The limitation of treatment was the risk of infection and malignancy. Intravenous anti-D showed some effectiveness in patients with acute and chronic ITP; however, this medication is unavailable in some countries. Objective: To determine the effectiveness and outcome of intramuscular anti-D in children with chronic ITP. Methods: Patients, children age≤18 years, diagnosed with chronic ITP with platelet count <20,000/uL, were enrolled. Treatment protocol, intramuscular anti-D (Igamed®) of 10 mcg/kg/dose, was divided into 3 phases of varied regimens: phase I - anti-D daily for 5 days; phase II - anti-D weekly for 12 weeks and withheld when platelet counts ≥20,000/uL; and phase III - anti-D once every 2 weeks for 24 weeks. Direct Coombs' test and LDH and hemoglobin (Hb) levels were monitored on day 3, 5 and 7 during phase I; every week during phase II and every 2 weeks during phase III. The response was defined by platelet count>20,000/uL. Results: Twenty-five patients with chronic ITP, median (range) age 7.8(3.8–15.5) years, were enrolled, 9 patients [median age 11.3(4.6–15.5) years, females:males = 2:7] were treated with anti-D while 16 patients [median age 6.6(3.8–12.3) years, females:males = 4:12] were not. Median platelet count at the time of enrollment was 7,000(2,000–18,000)/uL. Median platelet counts in anti-D treatment [9,000(2,000–18,000)/uL] and non-treatment [4,500(2,000–16,000)/uL] groups at the time of enrollment were not significantly different. During phase I, the median platelet counts in all treated patients significantly increased from 9,000(6,500–14,700)/uL to 32,000(22,430–46,000)/uL, p= 0.012. The maximum response was during days 4–5 after treatment. Seven patients received anti-D weekly in phase II and every 2 weeks in phase III with a total of 84 episodes each in both phases. The results revealed platelet count≥ 20,000/uL in 21 episodes [25(8.3–78.2)%] in phase II and 25.5 episodes [30.7(0–50)%] in phase III. Direct Coombs' tests were positive (3+ to 4+) in all patients; however, Hb level was not changed. The median follow-up time after enrollment in anti-D treatment group was 2.2(0.6–3.7) years. One patient in anti-D treatment group was in remission (platelet>100,000/uL) at 1.3 and 0.8 years after diagnosis and enrollment, respectively and 8 patients in non-treatment group were in remission at median time of 4.1(1.6–5.4) and 2.0(0.8–4.2) years after diagnosis and enrollment, respectively, p= 0.22. Conclusion: Intramuscular anti-D demonstrated the 100% response when given 10 mcg/kg/dose for 5 days. The once a week and once every 2 weeks showed only 25% and 30% response, respectively. Anti-D treatment during 37 weeks did not affect the long-term outcome. No complication was detected. Therefore, intramuscular anti-D given 10 mcg/kg for 5 days can be an alternative method to raise platelet count for immediate invasive procedure requirement in chronic ITP patients with severe thrombocytopenia. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 54 (12) ◽  
pp. 1252-1259
Author(s):  
Barbara A. Santevecchi ◽  
Stacy Miller ◽  
Lindsey M. Childs-Kean

Objective: To review data on efficacy and safety of dolutegravir (DTG) and lamivudine (3TC) in treatment-naïve adults with HIV-1 infection. Data Sources: Phase III clinical trials and review articles were identified through PubMed (1996 to March 2020) and ClinicalTrials.gov (2000 to May 2020) using the keywords dolutegravir, lamivudine, and HIV. Study Selection and Data Extraction: Relevant clinical trials and review articles available in English evaluating efficacy and safety of DTG and 3TC were included. Data Synthesis: The once-daily, single-tablet regimen of DTG/3TC is the first dual antiretroviral therapy (ART) recommended for initial therapy in treatment-naïve adults with HIV-1 infection. DTG and 3TC were compared with a regimen of DTG and tenofovir disoproxil fumarate/emtricitabine in the GEMINI studies and demonstrated noninferiority for the primary end point of virological suppression at up to 96 weeks. No treatment-emergent resistance mutations were identified in a small group of participants who did not reach virological suppression. The regimen is well tolerated, and the most common adverse events reported in trials include headache, diarrhea, nausea, insomnia, and fatigue. Relevance to Patient Care and Clinical Practice: This dual-ART regimen is a favorable treatment option for ART-naïve patients with HIV-1 RNA <500 000 copies/mL, absence of hepatitis B virus, and no resistance to DTG or 3TC. Benefits of dual ART include reduction in treatment-related adverse events and toxicities, drug interactions, and cost. In addition, the once-daily, single-tablet formulation promotes adherence. Conclusions: DTG/3TC has demonstrated efficacy in maintaining virological suppression in ART-naïve patients at up to 96 weeks while minimizing treatment-related adverse events and toxicities.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1307-1307
Author(s):  
Tadeusz Robak ◽  
Abdulgabar Salama ◽  
Lidia Kovaleva ◽  
Yaroslava I. Vyhovska ◽  
Simon Davies ◽  
...  

Abstract Background Intravenous immunoglobulin (IVIG) is an accepted treatment for immune thrombocytopenic purpura (ITP). A new liquid 10% human IgG preparation stabilized with proline at a pH of 4.8 (trade name: Privigen) was recently developed. It can be stored at room temperature and is therefore always ready to use. Here we report its efficacy and safety in patients with chronic ITP. Patients and Methods Fifty-seven patients with chronic ITP and a platelet count below 20 x 109/L were included in this open-label, single arm, multi-center Phase III trial. IVIG was given at a dose of 1 g/kg on 2 consecutive days at a maximum infusion rate of 4 mg/kg/min. A subset (56.1%) of the patients received premedication (acetaminophen or diphenhydramine) to avoid adverse events. The primary endpoint was the platelet response rate, defined as the percentage of patients showing an increase in platelet count to ≥50 x 109/L within 7 days of the first infusion. Secondary endpoints included platelet counts at specified time points, time to platelet response, duration of platelet response, and regression of hemorrhages at different bleeding sites. Safety was evaluated by the frequency and severity of adverse events. Results The primary endpoint, an increase in platelet counts to ≥50 x 109/L, was achieved by 81% of the subjects (95% CI: 69–89%). The highest median platelet count (149 x 109/L) was observed on day 8. Median time to response was 2.5 days, with 43% of subjects responding within 1 day. Median duration of platelet response (days with platelet count ≥ 50 x 109/L) was 15.4 days. Regression rates for various bleeding sites ranged from 78% to 100%. Regression of bleeding correlated with increases in platelet counts. Adverse events were reported in 52 (91%) subjects. The most common adverse event was headache (67%), the incidence and severity of which was attenuated by premedication with acetaminophen/diphenhydramine. There were 3 serious adverse events, one of which (aseptic meningitis) was considered related to the study medication. Conclusions The present study has shown the safety and significant efficacy of a novel, 10% liquid, ready for use IVIG preparation, in patients with chronic ITP. A rapid increase in platelet counts to a level where severe bleeding episodes become more unlikely was seen in the majority of patients, as expected with IVIG given at 1 g/kg on 2 consecutive days. The increase in platelet counts was associated with a regression of bleeding. Adverse events were generally mild to moderate in severity, corresponded to those expected with IVIG, and could be prevented with premedication.


Vaccines ◽  
2021 ◽  
Vol 9 (6) ◽  
pp. 582
Author(s):  
Haoyue Cheng ◽  
Zhicheng Peng ◽  
Wenliang Luo ◽  
Shuting Si ◽  
Minjia Mo ◽  
...  

Nowadays, the vaccination with COVID-19 vaccines is being promoted worldwide, professionals and common people are very concerned about the efficacy and safety of COVID-19 vaccines. No published systematic review and meta-analysis has assessed the efficacy and safety of the COVID-19 vaccines based on data from phase III clinical trials. Therefore, this study has estimated the efficacy and safety of COVID-19 vaccines and the differences between vaccine types. PubMed, Embase, the Cochrane Library, CNKI, Wanfang, medRxiv databases and two websites were used to retrieve the studies. Random-effects models were used to estimate the pooled efficacy and safety with risk ratio (RR). A total of eight studies, seven COVID-19 vaccines and 158,204 subjects were included in the meta-analysis. All the vaccines had a good preventive effect on COVID-19 (RR = 0.17, 95% CI: 0.09–0.32), and the mRNA vaccine (RR = 0.05, 95% CI: 0.03–0.09) was the most effective against COVID-19, while the inactivated vaccine (RR = 0.32, 95% CI: 0.19–0.54) was the least. In terms of safety, the risk of overall adverse events showed an increase in the vaccine group after the first (RR = 1.46, 95% CI: 1.03–2.05) or second (RR = 1.52, 95% CI: 1.04–2.20) injection. However, compared with the first injection, the risk of local (RR = 2.64, 95% CI: 1.02–6.83 vs. RR = 2.25, 95% CI: 0.52–9.75) and systemic (RR = 1.33, 95% CI: 1.21–1.46 vs. RR = 1.59, 95% CI: 0.84–3.01) adverse events decreased after the second injection. As for the mRNA vaccine, the risk of overall adverse events increased significantly, compared with the placebo, no matter whether it was the first (RR = 1.83, 95% CI = 1.80–1.86) or the second (RR = 2.16, 95% CI = 2.11–2.20) injection. All the COVID-19 vaccines that have published the data of phase III clinical trials have excellent efficacy, and the risk of adverse events is acceptable. The mRNA vaccines were the most effective against COVID-19, meanwhile the risk and grade of adverse events was minimal, compared to that of severe symptoms induced by COVID-19.


PLoS ONE ◽  
2021 ◽  
Vol 16 (3) ◽  
pp. e0244786
Author(s):  
Jew Ochola Ouma ◽  
David H. Mulama ◽  
Lucas Otieno ◽  
John Owuoth ◽  
Bernhards Ogutu ◽  
...  

There is an urgent need for reliable region-specific hematological reference values for clinical monitoring. Laboratory reference ranges are important for assessing study participant eligibility, toxicity grading and management of adverse events in clinical trials and clinical diagnosis. Most clinical laboratories in Kenya rely on hematological reference values provided by instrument manufacturers and/or textbooks, which are based on population from Europe or North America. The use of such values in medical practice could result in improper patient management, selection bias in selection of appropriate participants for clinical trials and flawed classification of the clinical adverse events when applied to African populations. The aim of this study was to establish local laboratory hematological reference values in infants aged 1 month to 17 months from Kombewa Sub-county that could be true representative of the existing rural population. The study participants in the current study were those who had previously been recruited from GSK-sponsored study. This study was a phase III, Double Blind, Randomized, GSK-sponsored, Malaria Vaccine Clinical Trial that was conducted in infants aged 1month to 17months. 1,509 participants were included in the study analysis. Data were partitioned into 3 different age groups (1–6 months[m], 6–12 m and 12–17 m) and differences between gender were compared within each group. Data were analyzed using Graphpad prism V5 to generate 95% reference ranges (2.5th-97.5th percentile). There was evidence of gender differences in hemoglobin values (p = 0.0189) and platelet counts (p = 0.0005) in the 1 to 6m group. For the 12-17m group, there were differences in MCV (p<0.0001) and MCH (p = 0.0003). Comparing gender differences for all age groups, differences were noted in percent lymphocytes (p = 0.0396), percent monocytes (p = 0.0479), percent granulocytes (p = 0.0044), hemoglobin (p = 0.0204), hematocrit (p = 0.0448), MCV (p = 0.0092), MCH (p = 0.0089), MCHC (p = 0.0336) and absolute granulocytes (p = 0.0237). In 1 to 6m age group and all age groups assessed, for WBCs, hemoglobin, hematocrit, MCV and lymphocytes absolute counts, both 2.5th and 97.5th percentiles for Kisumu infants were higher than those from Kilifi. Platelet ranges for Kisumu children were narrower compared to Kilifi ranges. Kisumu hematology reference ranges were observed to be higher than the ranges of Tanzanian children for the WBCs, absolute lymphocyte and monocyte counts, hemoglobin, hematocrit and MCV. Higher ranges of WBCs, absolute lymphocyte and monocyte counts were observed compared to the values in US/Europe. Wider ranges were observed in hemoglobin, hematocrit, and MCV. Wider ranges were observed in platelet counts in Kisumu infants compared to the US/Europe ranges. Compared to Harriet Lane Handbook reference values that are used in the area, higher counts were observed in WBC counts, both absolute and percent lymphocyte counts, as well as monocyte counts for current study. Wider ranges were observed in RBC, platelets and RDW, while lower ranges noted in the current study for hemoglobin, hematocrit and granulocyte counts. This study underscores the importance of using locally established hematology reference ranges of different age groups in support of proper patient management and for clinical trials.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 176-176 ◽  
Author(s):  
Moshe Talpaz ◽  
Ronald Paquette ◽  
Lawrence Afrin ◽  
Solomon Hamburg ◽  
Katarzyna Jamieson ◽  
...  

Abstract Abstract 176 Background: Ruxolitinib (RUX) has demonstrated clinical benefit for patients with myelofibrosis (MF) with or without the JAK2V617F mutation at starting doses of 15 or 20 mg PO BID by alleviating symptoms, improving quality of life measures, reducing spleen volume and exhibiting an apparent increase in overall survival in the phase III placebo (PBO)-controlled COMFORT-I study. Reversible declines in platelet count and hemoglobin (Hgb) can occur with ruxolitinib but are rarely treatment-limiting. Patients with MF who have low platelet counts represent an important subset of MF patients; given the potential risk of bleeding complications, a dosing strategy for such patients is needed. We assessed an alternative strategy using lower starting doses of ruxolitinib with subsequent dose escalation in patients with MF who have platelet counts of 50–100 × 109/L (Study INCB018424-258; NCT01348490). Methods: RUX dosing started at 5 mg BID. With adequate platelet count, doses could increase by 5 mg once daily every 4 weeks to 10 mg BID. Further increases required evidence of suboptimal efficacy. Assessments include measurement of MF symptoms (MF Symptom Assessment Form v2.0 Total Symptom Score [TSS]); Patient Global Impression of Change (PGIC); EORTC QLQ-C30, measurement of spleen volume by MRI, and safety/tolerability. Results: A total of 50 patients have enrolled, with data available for 41 patients. Nineteen have completed 24 weeks of treatment; >70% of these patients attained a final dose of ≥10 mg BID of RUX. Treatment was generally well tolerated in this study population with no withdrawals for thrombocytopenia or bleeding events. Based on analysis of adverse events, no new safety signals were observed in this population of MF patients with low platelet counts. Data for efficacy parameters, including spleen volume reduction, TSS reduction, and improvement in EORTC-QLQ-C30 subscales and PGIC were consistent with RUX treatment in the COMFORT-I study, and demonstrated clinically meaningful efficacy compared with the COMFORT-I PBO arm (Table). Of 19 patients with platelet count data through Week 24, 5 showed increased platelet count over the duration of the study (range of increase: 20 to 95 x109/L); all 5 patients had optimized dosing to ≥10 mg BID. Compared with the 14 patients showing smaller increases or modest decreases in platelet count, these 5 patients were younger (mean age: 63 years vs. 71 years), had been diagnosed with MF more recently (2.2 years vs. 5.2 years) and had lower DIPSS scores (60% Intermediate-1; 20% Intermediate-2; 20% High vs. 0% Intermediate-1; 79% Intermediate-2; 21% High). Four patients (9.8%) reported adverse events of bleeding (excluding events related to bruising) of any grade (all events were Grade 1 except one Grade 2 hematochezia), consistent with previously reported hemorrhage frequency in the COMFORT-I study (16.8%, RUX; 12.6%, PBO). Conclusions: These preliminary findings suggest that a dosing strategy of a low starting dose of RUX with escalation to 10 mg BID may be appropriate in MF patients who have low platelet counts. Most patients were able to titrate to a dose of ≥10 mg BID of RUX, a dose showing efficacy for both spleen volume and patient-reported outcomes generally consistent with previously reported data from Phase III trials. An increase in platelet counts was observed in approximately one-fourth of patients who completed 24 weeks of RUX treatment. Escalation to, and subsequent maintenance of, a 10 mg BID dose of RUX also preserves both Hgb and platelet count which may be beneficial for MF patients with anemia or thrombocytopenia. Disclosures: Talpaz: Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; BMS: Membership on an entity's Board of Directors or advisory committees, Research Funding; Ariad: Research Funding; Sanofi: Membership on an entity's Board of Directors or advisory committees, Research Funding; Teva: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees. Paquette:Incyte: Consultancy. Jamieson:Sunesis: Membership on an entity's Board of Directors or advisory committees; Blue Distinction Centers for Transplants BlueCross BlueShield Association: Consultancy. Lyons:Amgen: Consultancy, Research Funding; Incyte: Consultancy, Research Funding; Novartis: Research Funding. Tiu:Incyte: Honoraria, Speakers Bureau. Winton:Incyte Corporation: Consultancy, Honoraria. Odenike:Incyte: Membership on an entity's Board of Directors or advisory committees; Sanofi-Aventis:Membership on an entity's Board of Directors or advisory committees. Peng:Incyte: Employment, Equity Ownership. Sandor:Incyte: Employment, Equity Ownership. O'Neill:Incyte: Employment, Equity Ownership. Erickson-Viitanen:Incyte: Employment, Equity Ownership. Leopold:Incyte: Employment, Equity Ownership. Levy:Incyte: Employment, Equity Ownership. Kantarjian:Incyte Corporation: grant support Other. Verstovsek:Incyte Corporation: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document