Network


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

Hotspot


Dive into the research topics where Amy D. Shapiro is active.

Publication


Featured researches published by Amy D. Shapiro.


Haemophilia | 2003

Consensus perspectives on prophylactic therapy for haemophilia: summary statement

Erik Berntorp; Jan Astermark; Sven Björkman; Victor S. Blanchette; K. Fischer; Paul Giangrande; A. Gringeri; Rolf Ljung; Marilyn J. Manco-Johnson; M. Morfini; R. F. Kilcoyne; Pia Petrini; E. C. Rodriguez-Merchan; Wolfgang Schramm; Amy D. Shapiro; H. M. Van Den Berg; C. Hart

Summary.  Participants in an international conference on prophylactic therapy for severe haemophilia developed a consensus summary of the findings and conclusions of the conference. In the consensus, participants agreed upon revised definitions for primary and secondary prophylaxis and also made recommendations concerning the need for an international system of pharmacovigilance. Considerations on starting prophylaxis, monitoring outcomes, and individualizing treatment regimens were discussed. Several research questions were identified as needing further investigation, including when to start and when to stop prophylaxis, optimal dosing and dose interval, and methods for assessment of long‐term treatment effects. Such studies should include carefully defined cohorts, validated orthopaedic and quality‐of‐life assessment instruments, and cost‐benefit analyses.


Blood | 2014

Phase 3 study of recombinant factor VIII Fc fusion protein in severe hemophilia A

Johnny Mahlangu; Jerry S. Powell; Margaret V. Ragni; Pratima Chowdary; Neil C. Josephson; Ingrid Pabinger; Hideji Hanabusa; Naresh Gupta; Roshni Kulkarni; Patrick F. Fogarty; David J. Perry; Amy D. Shapiro; K. John Pasi; Shashikant Apte; Ivan Nestorov; Haiyan Jiang; Shuanglian Li; Srividya Neelakantan; Lynda M. Cristiano; Jaya Goyal; Jurg M. Sommer; Jennifer A. Dumont; Nigel Dodd; Karen Nugent; Gloria Vigliani; Alvin Luk; Aoife Brennan; Glenn F. Pierce

This phase 3 pivotal study evaluated the safety, efficacy, and pharmacokinetics of a recombinant FVIII Fc fusion protein (rFVIIIFc) for prophylaxis, treatment of acute bleeding, and perioperative hemostatic control in 165 previously treated males aged ≥12 years with severe hemophilia A. The study had 3 treatment arms: arm 1, individualized prophylaxis (25-65 IU/kg every 3-5 days, n = 118); arm 2, weekly prophylaxis (65 IU/kg, n = 24); and arm 3, episodic treatment (10-50 IU/kg, n = 23). A subgroup compared recombinant FVIII (rFVIII) and rFVIIIFc pharmacokinetics. End points included annualized bleeding rate (ABR), inhibitor development, and adverse events. The terminal half-life of rFVIIIFc (19.0 hours) was extended 1.5-fold vs rFVIII (12.4 hours; P < .001). Median ABRs observed in arms 1, 2, and 3 were 1.6, 3.6, and 33.6, respectively. In arm 1, the median weekly dose was 77.9 IU/kg; approximately 30% of subjects achieved a 5-day dosing interval (last 3 months on study). Across arms, 87.3% of bleeding episodes resolved with 1 injection. Adverse events were consistent with those expected in this population; no subjects developed inhibitors. rFVIIIFc was well-tolerated, had a prolonged half-life compared with rFVIII, and resulted in low ABRs when dosed prophylactically 1 to 2 times per week.


Blood | 2012

Recombinant factor IX-Fc fusion protein (rFIXFc) demonstrates safety and prolonged activity in a phase 1/2a study in hemophilia B patients

Amy D. Shapiro; Margaret V. Ragni; Leonard A. Valentino; Nigel S. Key; Neil C. Josephson; Jerry S. Powell; Gregory Cheng; Arthur R. Thompson; Jaya Goyal; Karen L. Tubridy; Robert T. Peters; Jennifer A. Dumont; Donald Euwart; Lian Li; Bengt Hallén; Peter Gozzi; Alan J. Bitonti; Haiyan Jiang; Alvin Luk; Glenn F. Pierce

Current factor IX (FIX) products display a half-life (t(1/2)) of ∼ 18 hours, requiring frequent intravenous infusions for prophylaxis and treatment in patients with hemophilia B. This open-label, dose-escalation trial in previously treated adult subjects with hemophilia B examined the safety and pharmacokinetics of rFIXFc. rFIXFc is a recombinant fusion protein composed of FIX and the Fc domain of human IgG(1), to extend circulating time. Fourteen subjects received a single dose of rFIXFc; 1 subject each received 1, 5, 12.5, or 25 IU/kg, and 5 subjects each received 50 or 100 IU/kg. rFIXFc was well tolerated, and most adverse events were mild or moderate in intensity. No inhibitors were detected in any subject. Dose-proportional increases in rFIXFc activity and Ag exposure were observed. With baseline subtraction, mean activity terminal t(1/2) and mean residence time for rFIXFc were 56.7 and 71.8 hours, respectively. This is ∼ 3-fold longer than that reported for current rFIX products. The incremental recovery of rFIXFc was 0.93 IU/dL per IU/kg, similar to plasma-derived FIX. These results show that rFIXFc may offer a viable therapeutic approach to achieve prolonged hemostatic protection and less frequent dosing in patients with hemophilia B. The trial was registered at www.clinicaltrials.gov as NCT00716716.


Journal of Pediatric Hematology Oncology | 1997

Factor IX inhibitors and anaphylaxis in hemophilia B

Indira Warrier; Bruce M. Ewenstein; Marion A. Koerper; Amy D. Shapiro; Nigel S. Key; Donna DiMichele; Robert T. Miller; John Pasi; Georges E. Rivard; Steve S. Sommer; Jacob Katz; Frauke Bergmann; Rolf Ljung; Pia Petrini; Jeanne M. Lusher

PURPOSE We present clinical and laboratory data on 18 children from 12 hemophilia treatment centers in the United States, Canada, and Europe with the purpose of disseminating information regarding a recently recognized, potentially life-threatening complication of treatment in very young children with hemophilia B. PATIENTS AND METHODS Twelve hemophilia centers from the United States, Canada, and Europe provided clinical information and laboratory data concerning 18 children who had severe allergic reactions to infused factor (F) IX in close association with the development of an inhibitor to FIX. Laboratory testing for establishment of the diagnosis of hemophilia B and inhibitor to FIX was done locally at the centers treating these patients. FIX gene analysis was performed at one of six molecular genetics institutes. RESULTS All 18 children had severe hemophilia B, and in each an inhibitor antibody to FIX developed. The median age at the time of anaphylaxis (or anaphylactoid reaction) was 16 months, and the median number of exposure days to FIX was 11. The FIX inhibitor was detected almost simultaneously with the first occurrence of anaphylaxis in 12 of 18 patients. Maximum inhibitor titers were 4.5-600 Bethesda units (BU), with a median titer of 48 BU. FIX gene analysis, performed in 17 of 18 patients, demonstrated complete deletion of the FIX gene in 10 and major derangements in seven. Immune tolerance induction (ITI) regimens have been attempted in 12 patients, with generally poor responses. Two of the 12 experienced nephrotic syndrome while on ITI. Recombinant FVIIa has been successfully used to treat bleeding episodes in 11 of these children. CONCLUSION Physicians treating young children with hemophilia B should be aware of the potentially life-threatening complication of anaphylaxis. Children with complete gene deletions or major derangements of the FIX gene appear to be at greater risk. Those identified by genotype as being at greater risk may need to receive their first 10-20 treatments in a medical facility equipped for handling such emergencies. Recombinant FVIIa, although not licensed for use in the United States, appears to be the most suitable treatment option for bleeding episodes in such patients.


The Lancet | 2012

Modern haemophilia care.

Erik Berntorp; Amy D. Shapiro

Haemophilia care has undergone substantial improvements during the past 40-50 years. Early clotting factor concentrates were not sufficiently refined to enable self-administered treatment at home until the 1970s. Unfortunately, these advances led to transmission of viral diseases including HIV and hepatitis, resulting in an increased burden of morbidity and mortality, especially during the 1980s. Throughout the past two decades, product development, including the advent of recombinant concentrates, has greatly improved the safety and availability of therapy and the focus of care is shifting towards prevention and management of disease sequelae. Long-term substitution therapy (prophylaxis) of the missing clotting factor is the recommended treatment in severe haemophilia, but several research issues remain to be elucidated such as when to start and how to optimise these regimens, and when or whether to stop this expensive treatment. The major side-effect of treatment, development of inhibitors to the infused concentrate, is the main threat to the health of patients and consequently the goal of intense research. Development of new products with improved pharmacokinetics is the next step to improved therapy.


Haemophilia | 2005

Dose effect and efficacy of rFVIIa in the treatment of haemophilia patients with inhibitors: analysis from the Hemophilia and Thrombosis Research Society Registry

R. Parameswaran; Amy D. Shapiro; Joan Cox Gill; Craig M. Kessler; Thomas C. Abshire; Anne L. Angiolillo; Lisa N. Boggio; A. Cohen; Donna DiMichele; W. Hanna; Keith Hoots; J. Hord; Nigel S. Key; Barbara A. Konkle; Peter A. Kouides; E. Kurczynski; P. Marks; Joseph E. Palascak; S. Pipe; Margaret V. Ragni; G. Rivard; R. Shopnick; Michael Tarantino; Leonard A. Valentino; R. Watts; Gilbert C. White

Summary.  Recombinant activated factor VII (rFVIIa), licensed in 1999 for treatment of haemophilia patients with inhibitors (HI), represents an important advance in the therapeutic armamentarium. Standard bolus dosing ranges from 90 to 120 mcg kg−1 every 2–3 h until arrest of bleeding. As licensure, clinical use of rFVIIa has increased and broadened. Clinicians now use a wide dose range, 90–300 mcg kg−1. High‐dose regimens may optimize thrombin generation or burst, and may allow for prolonged dose interval. The Hemophilia and Thrombosis Research Society (HTRS) maintains a registry database to study haemophilia treatment and related disorders, particularly treatment of acute bleeding in HI, acquired haemophilia, FVII deficiency and von Willebrands disease (VWD). To assess the effect of rFVIIa dose on efficacy and safety in the treatment of acute bleeding in HI, data from the HTRS database from January 2000 through June 2002 were analysed. Bleeding episodes were grouped by bolus rFVIIa dose range: <100, 100–150, 150–200 and >200 mcg kg−1. Investigator‐reported efficacy for the first 72 h of treatment was evaluated. Thirty‐eight congenital HI patients were treated for 555 bleeding episodes. Patient age range was 1–55 years (median: 14). Bleeding episodes were spontaneous (45%), caused by trauma (38%), or because of surgery, dental, diagnostic, or medical procedures (17%); bleeding occurred in joint, muscle, and intra/extracranial sites. Treatment location included: 80% at home, 12% at other facilities (treatment centres, ER, inpatient and OR), and 8% at both home/other facilities. Median total dose given over 72 h was 360 mcg kg−1 (range: 40–4281, mean: 537). Bleeding stopped in 87% of the episodes. Bleeding cessation rate was 84% for the three lower dose groups, and 97% for the highest dose group (P < 0.001). Five patients experienced nine adverse events (AEs). AE rates were <1% for <100, 5% for 100–150, 0% for 150–200, <1% for >200 mcg kg−1 dose group. Decreased therapeutic response accounted for eight of the nine AEs. These data, which represent the most comprehensive report of rFVIIa use since the USA licensure, demonstrate that bleeding episodes in HI patients can be treated safely and effectively at home and that doses up to 346 mcg kg−1 appear to be well‐tolerated. Additionally, rFVIIa doses >200 mcg kg−1 appear to significantly increase efficacy (97% in the high‐dose group, compared with 84% in the lower dose groups). Optimal dosing remains to be determined; specifically, what the lowest effective dose is and whether a single high‐dose bolus eliminates the need for repeated dosing. Recombinant FVIIa appears to have a wide safety margin that may allow dose escalation to address these questions.


Haemophilia | 2004

Clinical evaluation of an advanced category antihaemophilic factor prepared using a plasma/albumin-free method: pharmacokinetics, efficacy, and safety in previously treated patients with haemophilia A

Michael Tarantino; Peter William Collins; C. R. M. Hay; Amy D. Shapiro; Ralph A. Gruppo; Erik Berntorp; G. L. Bray; S. A. Tonetta; Phillip Schroth; A. D. Retzios; S. S. Rogy; M. G. Sensel; Bruce M. Ewenstein

Summary.  The efficacy and safety of an advanced category recombinant antihaemophilic factor produced by a plasma‐ and albumin‐free method (rAHF‐PFM) was studied in 111 previously treated subjects with haemophilia A. The study comprised a randomized, double‐blinded, crossover pharmacokinetic comparison of rAHF‐PFM and RECOMBINATE rAHF (R‐FVIII); prophylaxis (three to four times per week with 25–40 IU kg−1 rAHF‐PFM) for at least 75 exposure days; and treatment of episodic haemorrhagic events. Median age was 18 years, 96% of subjects had baseline factor VIII <1%, and 108 received study drug. Bioequivalence, based on area under the plasma concentration vs. time curve and adjusted in vivo recovery, was demonstrated for rAHF‐PFM and R‐FVIII. Mean (±SD) half‐life for rAHF‐PFM was 12.0 ± 4.3 h. Among 510 bleeding events, 473 (93%) were managed with one or two infusions of rAHF‐PFM and 439 (86%) had efficacy ratings of excellent or good. Subjects who were less adherent to the prophylactic regimen had a higher bleeding rate (9.9 episodes subject−1 year−1) than subjects who were more adherent (4.4 episodes subject−1 year−1; P < 0.03). One subject developed a low titre, non‐persistent inhibitor (2.0 BU) after 26 exposure days. These data demonstrate that rAHF‐PFM is bioequivalent to R‐FVIII, and suggest that rAHF‐PFM is efficacious and safe, without increased immunogenicity, for the treatment of haemophilia A.


Journal of Thrombosis and Haemostasis | 2008

Plasma and albumin-free recombinant factor VIII: pharmacokinetics, efficacy and safety in previously treated pediatric patients.

Victor S. Blanchette; A. D. Shapiro; R. J. Liesner; F Hernández Navarro; I. Warrier; Phillip Schroth; Gerald Spotts; Bruce M. Ewenstein; Thomas C. Abshire; A. Angiolillo; S. Arkin; David L. Becton; V. Blanchette; Alexis A. Thompson; Donna DiMichele; J. DiPaola; Keith Hoots; Margaret Heisel Kurth; C. Manno; I. Ortiz; Steven W. Pipe; Michael Recht; F. Shafer; Amy D. Shapiro; Michael Tarantino; W. Y. Wong; Christoph Male; M. Siimes; Thierry Lambert; Chantal Rothschild

Summary.  Background: The pharmacokinetics of factor VIII replacement therapy in preschool previously treated patients (PTPs) with hemophilia A have not been well characterized. Objectives: To assess the pharmacokinetics, efficacy and safety of a plasma‐free recombinant FVIII concentrate, ADVATE [Antihemophilic Factor (Recombinant), Plasma/Albumin‐Free Method, rAHF‐PFM], in children < 6 years of age with severe hemophilia. Patients/methods: Fifty‐two boys, one girl, mean (± SD) age 3.1 ± 1.5 years and ≥ 50 days of prior FVIII exposure, were enrolled in a prospective study of ADVATE rAHF‐PFM at 23 centers. Results: The mean terminal phase half‐life (t1/2) was 9.88 ± 1.89 h, and the mean adjusted in vivo recovery (IVR) was 1.90 ± 0.43 IU dL−1 (IU kg−1)−1. Over the 1–6‐year age range, t1/2 of rAHF‐PFM increased by 0.40 h year−1. IVR increased by 0.095IU dL−1(IU kg−1)−1 (kg m−2)−1 in relation to body mass index (BMI). Patients primarily received prophylaxis. Median (range) annual joint bleeds were 0.0 (0.0–5.8), 0.0 (0.0–6.1) and 14.2 (0.0–34.5) for standard prophylaxis, modified prophylaxis and on‐demand treatment, respectively. Bleeds were managed in 90% (319/354) of episodes with one or two rAHF‐PFM infusions; response was rated excellent/good in 93.8% of episodes. Over a median 156 exposure days, no FVIII inhibitors were detected and no related severe adverse events or unusual non‐serious adverse events were seen. Conclusions: Children < 6 years of age appear to have shorter FVIII t1/2 and lower IVR values than older subjects. However, these parameters increased with age (t1/2) and BMI (adjusted IVR), respectively. rAHF‐PFM was clinically effective and well tolerated, with no signs of increased immunogenicity in previously treated young children with hemophilia A.


Transfusion | 2002

Pharmacokinetic analysis of plasma-derived and recombinant F IX concentrates in previously treated patients with moderate or severe hemophilia B

Bruce M. Ewenstein; J. Heinrich Joist; Amy D. Shapiro; Thomas Hofstra; Cindy Leissinger; Stephanie V. Seremetis; Martin Broder; Guenther Mueller‐Velten; Bruce A. Schwartz

BACKGROUND: Hemophilia B is an X‐linked bleeding disorder that affects approximately 1 in 25,000 males. Therapy for acute bleeding episodes consists of transfusions of plasma‐derived (pd‐F IX) or recombinant (r‐F IX) concentrates.


Haemophilia | 2001

Pharmacokinetics of recombinant factor IX in relation to age of the patient: implications for dosing in prophylaxis

Sven Björkman; Amy D. Shapiro; Erik Berntorp

The aims of this study were to investigate possible age‐related changes in the disposition of factor IX procoagulant activity (FIX:C) after administration of recombinant factor IX (rFIX) and to translate the pharmacokinetic findings into suggestions for dosing of rFIX during prophylactic treatment of haemophilia B. Pharmacokinetic data were available from a previous study on 56 patients, aged 4–56 years (one of whom was excluded from analysis). FIX:C curves during prophylactic dosing were computer‐simulated from the single‐dose data. Clearance and volume of distribution at steady state of FIX:C increased linearly with body weight of the patients, consequently increasing during childhood and adolescence but remaining fairly constant during adulthood. The terminal half‐life of FIX:C showed no correlation with age, while in vivo recovery (in U dL–1 per U kg–1 given) tended to increase. Computer‐predicted trough levels of exogenous FIX:C during repeated doses of rFIX (50 U kg–1) and, conversely, doses (in U kg–1) needed to maintain a 1‐U dL–1 trough level showed little or no dependence on age. There was considerable interindividual variation in disposition and required doses of rFIX, emphasizing the need for individual dose titration. Dosing of rFIX according to lean body mass instead of body weight did not reduce this variability. During prophylaxis a 1‐U dL–1 trough level can normally be maintained by dosing every 2–3 days, the former schedule resulting in, on average, a 45% lower consumption of rFIX.

Collaboration


Dive into the Amy D. Shapiro's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joan Cox Gill

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Thomas C. Abshire

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael Tarantino

University of Illinois at Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge