Network


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

Hotspot


Dive into the research topics where Surya Ayalasomayajula is active.

Publication


Featured researches published by Surya Ayalasomayajula.


British Journal of Clinical Pharmacology | 2016

The effect of LCZ696 (sacubitril/valsartan) on amyloid-β concentrations in cerebrospinal fluid in healthy subjects

Thomas Langenickel; Chiaki Tsubouchi; Surya Ayalasomayajula; Parasar Pal; Marie-Anne Valentin; Markus Hinder; Stanford Jhee; Hakop Gevorkyan; Iris Rajman

Aims LCZ696 (angiotensin receptor neprilysin inhibitor) is a novel drug developed for the treatment of heart failure with reduced ejection fraction. Neprilysin is one of multiple enzymes degrading amyloid‐β (Aβ). Its inhibition may increase Aβ levels. The potential exists that treatment of LCZ696, through the inhibition of neprilysin by LBQ657 (an LCZ696 metabolite), may result in accumulation of Aβ. The aim of this study was to assess the blood–brain‐barrier penetration of LBQ657 and the potential effects of LCZ696 on cerebrospinal fluid (CSF) concentrations of Aβ isoforms in healthy human volunteers. Methods In a double‐blind, randomized, parallel group, placebo‐controlled study, healthy subjects received once daily LCZ696 (400 mg, n = 21) or placebo (n = 22) for 14 days. Results LCZ696 had no significant effect on CSF AUEC(0,36 h) of the aggregable Aβ species 1–42 or 1–40 compared with placebo (estimated treatment ratios 0.98 [95% CI 0.73, 1.34; P = 0.919] and 1.05 [95% CI 0.82, 1.34; P = 0.702], respectively). A 42% increase in CSF AUEC(0,36 h) of soluble Aβ 1–38 was observed (estimated treatment ratio 1.42 [95% CI 1.05, 1.91; P = 0.023]). CSF levels of LBQ657 and CSF Aβ 1–42, 1–40, and 1–38 concentrations were not related (r 2 values 0.022, 0.010, and 0.008, respectively). Conclusions LCZ696 did not cause changes in CSF levels of aggregable Aβ isoforms (1–42 and 1–40) compared with placebo, despite achieving CSF concentrations of LBQ657 sufficient to inhibit neprilysin. The clinical relevance of the increase in soluble CSF Aβ 1–38 is currently unknown.


British Journal of Clinical Pharmacology | 2015

The effect of LCZ696 on amyloid‐β concentrations in cerebrospinal fluid in healthy subjects

Thomas Langenickel; Chiaki Tsubouchi; Surya Ayalasomayajula; Parasar Pal; Marie-Anne Valentin; Markus Hinder; Stanford Jhee; Hakop Gevorkyan; Iris Rajman

Aims LCZ696 (angiotensin receptor neprilysin inhibitor) is a novel drug developed for the treatment of heart failure with reduced ejection fraction. Neprilysin is one of multiple enzymes degrading amyloid‐β (Aβ). Its inhibition may increase Aβ levels. The potential exists that treatment of LCZ696, through the inhibition of neprilysin by LBQ657 (an LCZ696 metabolite), may result in accumulation of Aβ. The aim of this study was to assess the blood–brain‐barrier penetration of LBQ657 and the potential effects of LCZ696 on cerebrospinal fluid (CSF) concentrations of Aβ isoforms in healthy human volunteers. Methods In a double‐blind, randomized, parallel group, placebo‐controlled study, healthy subjects received once daily LCZ696 (400 mg, n = 21) or placebo (n = 22) for 14 days. Results LCZ696 had no significant effect on CSF AUEC(0,36 h) of the aggregable Aβ species 1–42 or 1–40 compared with placebo (estimated treatment ratios 0.98 [95% CI 0.73, 1.34; P = 0.919] and 1.05 [95% CI 0.82, 1.34; P = 0.702], respectively). A 42% increase in CSF AUEC(0,36 h) of soluble Aβ 1–38 was observed (estimated treatment ratio 1.42 [95% CI 1.05, 1.91; P = 0.023]). CSF levels of LBQ657 and CSF Aβ 1–42, 1–40, and 1–38 concentrations were not related (r 2 values 0.022, 0.010, and 0.008, respectively). Conclusions LCZ696 did not cause changes in CSF levels of aggregable Aβ isoforms (1–42 and 1–40) compared with placebo, despite achieving CSF concentrations of LBQ657 sufficient to inhibit neprilysin. The clinical relevance of the increase in soluble CSF Aβ 1–38 is currently unknown.


Current Medical Research and Opinion | 2007

Evaluation of the potential for steady-state pharmacokinetic interaction between vildagliptin and simvastatin in healthy subjects

Surya Ayalasomayajula; Kiran Dole; Yan-Ling He; Monica Ligueros-Saylan; Yibin Wang; Joelle Campestrini; Henri Humbert; Gangadhar Sunkara

ABSTRACT Background: Vildagliptin is an orally active, potent and selective inhibitor of dipeptidyl peptidase IV (DPP-4), the enzyme responsible for the degradation of incretin hormones. By enhancing prandial levels of incretin hormones, vildagliptin improves glycemic control in type 2 diabetes. Co-administration of vildagliptin and simvastatin, an HMG-CoA-reductase inhibitor may be required to treat patients with diabetes and dyslipidemia. Therefore, this study was conducted to determine the potential for pharmacokinetic drug–drug interaction between vildagliptin and simvastatin at steady-state. Methods: An open label, single center, multiple dose, three period, crossover study was conducted in 24 healthy subjects. All subjects received once daily doses of either vildagliptin 100 mg or simvastatin 80 mg or the combination for 7 days with an inter-period washout of 7 days. Plasma levels of vildagliptin, simvastatin, and its active metabolite, simvastatin β-hydroxy acid (major active metabolite of simvastatin) were determined using validated LC/MS/MS methods. Pharmacokinetic and statistical analyses were performed using WinNonlin and SAS, respectively. Results: The 90% confidence intervals of Cmax and AUCτ of vildagliptin, simvastatin, and simvastatin β-hydroxy acid were between 80 and 125% (bioequivalence range) when vildagliptin and simvastatin were administered alone and in combination. These data indicate that the rate and extent of absorption of vildagliptin and simvastatin were not affected when co-administered, nor was the metabolic conversion of simvastatin to its active metabolite. All treatments were safe and well tolerated in this study. Conclusions: The pharmacokinetics of vildagliptin, simvastatin, and its active metabolite were not altered when vildagliptin and simvastatin were co-administered.


Current Medical Research and Opinion | 2008

A study of the pharmacokinetic interactions of the direct renin inhibitor aliskiren with allopurinol, celecoxib and cimetidine in healthy subjects

Surya Ayalasomayajula; Stéphanie Tchaloyan; Ching-Ming Yeh; Marie-Noelle Bizot; Hans Armin Dieterich; Dan Howard; William P. Dole

ABSTRACT Objective: Aliskiren is the first in a new class of orally effective direct renin inhibitors approved for the treatment of hypertension. This multiple-dose study investigated the potential for pharmacokinetic interactions between aliskiren and three drugs, each predominantly eliminated by a different clearance/metabolic pathway: allopurinol (glomerular filtration), celecoxib (cytochrome P450 metabolism) and cimetidine (P-glycoprotein and organic anion/cation transporters). Research design and methods: Three open-label, multiple-dose studies in healthy subjects investigated possible pharmacokinetic interactions between aliskiren 300 mg od and allopurinol 300 mg od (n = 20), celecoxib 200 mg bid (n = 22), or cimetidine 800 mg od (n = 22). Subjects received aliskiren alone or co-administered with allopurinol, celecoxib or cimetidine. Allopurinol and celecoxib were also administered alone and in combination with aliskiren. Plasma drug concentrations were determined by LC/MS/MS. Results: Co-administration of aliskiren with allopurinol had no effect on allopurinol AUCτ (ratio of geometric means 0.93 [90 % CI, 0.88, 0.98]) or oxypurinol AUCτ (mean ratio 1.12 [90 % CI, 1.08, 1.16]) and Cmax (mean ratio 1.08 [90 % CI, 1.04, 1.13]), with 90 % CI within the bioequivalence range 0.80–1.25, and a minor effect on allopurinol Cmax (mean ratio 0.88 [90 % CI, 0.78, 1.00]). Aliskiren co-administration had no effect on AUCτ or Cmax of celecoxib (mean ratios and 90 % CI within range 0.80–1.25). Neither allopurinol nor celecoxib significantly altered aliskiren AUCτ or Cmax (geometric mean ratios 0.88–1.02 with 90 % CI including 1.00, but with some 90 % CI outside the 0.80–1.25 range due to high variability). Co-administration of aliskiren with cimetidine increased aliskiren AUCτ by 20 % (mean ratio 1.20 [90 % CI, 1.07, 1.34]) and Cmax by 25 % (mean ratio 1.25 [90 % CI, 0.98, 1.59]). Conclusions: In this multiple-dose study, aliskiren showed no clinically relevant pharmacokinetic interactions when co-administered with allopurinol, celecoxib or cimetidine in healthy subjects.


Cardiovascular Therapeutics | 2016

Pharmacodynamic and Pharmacokinetic Profiles of Sacubitril/Valsartan (LCZ696) in Patients with Heart Failure and Reduced Ejection Fraction

Z. Kobalava; Y. Kotovskaya; Oleg Averkov; Elena Pavlikova; Valentine S. Moiseev; Diego Albrecht; Priya Chandra; Surya Ayalasomayajula; Margaret F. Prescott; Parasar Pal; Thomas Langenickel; Pierre Jordaan; Iris Rajman

Summary Aims Concomitant renin–angiotensin–aldosterone system blockade and natriuretic peptide system enhancement may provide unique therapeutic benefits to patients with heart failure and reduced ejection fraction (HFrEF). This study assessed the pharmacodynamics and pharmacokinetics of LCZ696 in patients with HFrEF. Methods This was an open‐label, noncontrolled single‐sequence study. After a 24‐h run‐in period, patients (n = 30) with HFrEF (EF ≤ 40%; NYHA class II–IV) received LCZ696 100 mg twice daily (bid) for 7 days and 200 mg bid for 14 days, along with standard treatment for heart failure (HF) (except angiotensin‐converting enzyme inhibitors [ACEIs] or angiotensin receptor blockers [ARBs]). Results On Day 21, significant increases were observed in the plasma biomarkers indicative of neprilysin and RAAS inhibition (ratio‐to‐baseline: cyclic guanosine monophosphate [cGMP], 1.38; renin concentration and activity, 3.50 and 2.27, respectively; all, P < 0.05). Plasma NT‐proBNP levels significantly decreased at all the time points on Days 7 and 21; plasma aldosterone and endothelin‐1 levels significantly decreased on Day 21 (all, P < 0.05). Following administration of LCZ696, the Cmax of sacubitril (neprilysin inhibitor prodrug), LBQ657 (active neprilysin inhibitor), and valsartan were reached within 0.5, 2.5, and 2 h. Between 100‐ and 200‐mg doses, the Cmax and AUC 0–12 h for sacubitril and LBQ657 were approximately dose‐proportional while that of valsartan was less than dose‐proportional. Conclusions Treatment with LCZ696 for 21 days was well tolerated and resulted in plasma biomarker changes indicative of neprilysin and RAAS inhibition in patients with HF. The pharmacokinetic exposure of the LCZ696 analytes in patients with HF observed in this study is comparable to that observed in the pivotal Phase III study.


The Journal of Clinical Pharmacology | 2008

Effects of Aliskiren, a Direct Renin Inhibitor, on Cardiac Repolarization and Conduction in Healthy Subjects

Surya Ayalasomayajula; Ching-Ming Yeh; Sujata Vaidyanathan; Brian Flannery; Hans Armin Dieterich; Dan Howard; Martin P. Bedigian; William P. Dole

This multicenter, double‐blind study evaluated the effects of aliskiren, a direct renin inhibitor approved for hypertension, on cardiac repolarization and conduction. Healthy volunteers (n = 298) were randomized to aliskiren 300 mg, aliskiren 1200 mg, moxifloxacin 400 mg (positive control), or placebo once daily for 7 days. Digitized electrocardiograms were obtained at baseline and day 7 of treatment over 23 hours postdose. Placebo‐adjusted mean changes from baseline in QTcF (Fridericia corrected), QTcI (individualized correction), PR, and QRS intervals were compared at each time point (time‐matched analysis) and for values averaged across the dosing period (baseline‐averaged analysis). In time‐matched analysis, mean changes in QTcF with aliskiren were below predefined limits for QTc prolongation (mean increase <5 milliseconds; upper 90% confidence interval [CI] <1 0 milliseconds) except aliskiren 1200 mg at 23 hours (5.2 milliseconds; 90% CI 2.2, 8.1). With moxifloxacin, significant QTcF prolongation occurred at most time points, confirming the sensitivity of the assay. Baseline‐averaged analysis was consistent with time‐matched analysis. Instances of QTcF interval >450 milliseconds or a >30‐millisecond increase from baseline with aliskiren (≤1%) were similar or lower than placebo (≤4%). Results were similar for QTcI. Aliskiren had no effect on PR or QRS duration. In conclusion, aliskiren at the highest approved dose (300 mg) and a 4‐fold higher dose had no effect on cardiac repolarization or conduction in healthy volunteers.


The Journal of Clinical Pharmacology | 2007

Effect of clopidogrel on the steady-state pharmacokinetics of fluvastatin.

Surya Ayalasomayajula; Sujata Vaidyanathan; Charisse Kemp; Pratapa Prasad; Alfred Balch; William P. Dole

This study assessed the effects of clopidogrel, a CYP 2C9 inhibitor, on fluvastatin pharmacokinetics in healthy volunteers. The effects of combined clopidogrel‐fluvastatin treatment on platelet function were also determined. Subjects received 80 mg fluvastatin (extended‐release formulation) alone on days 1 through 9, 80 mg fluvastatin and 300 mg clopidogrel (loading dose) on day 10, and 80 mg fluvastatin and 75 mg clopidogrel (maintenance dose) on days 11 through 19. Compared to treatment with fluvastatin alone, fluvastatin AUC was similar and Cmax increased marginally (15.7%) with concomitant treatment with clopidogrel. Platelet aggregation was inhibited by clopidogrel by 33% two hours after the loading dose and by 47% at steady state, similar to that reported for clopidogrel alone treatment. The authors conclude that coadministration of fluvastatin and clopidogrel has no clinically relevant effect on fluvastatin pharmacokinetics or on platelet inhibition by clopidogrel.


The Journal of Clinical Pharmacology | 2011

Evaluation of Pharmacokinetic Interactions Between Amlodipine, Valsartan, and Hydrochlorothiazide in Patients With Hypertension

Prafulla Bhad; Surya Ayalasomayajula; Rajesh Karan; Selene Leon; Gillies‐Jacques Riviere; Gangadhar Sunkara; Venkateswar Jarugula

The steady‐state pharmacokinetic (PK) interaction potential between amlodipine (10 mg), valsartan (320 mg), and hydrochlorothiazide (HCTZ; 25 mg) was evaluated in patients with hypertension in a multicenter, multiple‐dose, open‐label, 4‐cohort, parallel‐group study. Eligible patients were randomly allocated to the dual combination of valsartan + HCTZ, amlodipine + valsartan, or amlodipine + HCTZ and nonrandomly allotted to amlodipine + valsartan + HCTZ triple combination treatment. After 6 days of treatment with a half‐maximal dose of different combinations, patients were up‐titrated to the maximal drug doses from day 7 through day 17. PK parameters of corresponding analytes from the triple‐ and dual‐treatment groups were estimated on day 17 and compared. Safety and tolerability of all treatments was assessed. The Cssmax and AUC0‐τ values of amlodipine or HCTZ remained unaffected when administered with valsartan + HCTZ or valsartan + amlodipine, respectively. On the other hand, valsartan exposure increased by 10% to 25% when coadministered with HCTZ and amlodipine, which is not considered clinically relevant. In conclusion, there were no clinically relevant PK interactions with amlodipine, valsartan, and HCTZ triple combination compared with the corresponding dual combinations. All treatments were safe and well tolerated.


Clinical pharmacology in drug development | 2014

Effect of food on the oral bioavailability of amlodipine/valsartan and amlodipine/valsartan/hydrochlorothiazide fixed dose combination tablets in healthy subjects

Gangadhar Sunkara; Xuemin Jiang; Christine Reynolds; Denise Serra; Yiming Zhang; Monica Ligueros-Saylan; Surya Ayalasomayajula; Serge Winter; Venkateswar Jarugula

A double fixed dose combination of amlodipine/valsartan and triple fixed dose combination of amlodipine/valsartan/HCTZ tablets have been developed to treat patients with moderate‐to‐severe hypertension. Here, we present the effect of food on the oral bioavailability of these two fixed dose combination tablets from two separate clinical studies in healthy subjects. Single oral doses of amlodipine/valsartan (10/160 mg) and amlodipine/valsartan/HCTZ (10/320/25 mg were administered under fasted or fed conditions. Blood samples were collected in both studies to determine the pharmacokinetic parameters of amlodipine, valsartan, and/or HCTZ using non‐compartmental analysis. Following amlodipine/valsartan administration, the geometric mean ratios (GMRs, 90% CI) of AUC0–∞ and Cmax were 1.09 (1.05–1.13) and 1.03 (0.97–1.09) for amlodipine, and 0.94 (0.81–1.10) and 0.86 (0.73–1.02) for valsartan, respectively. Following amlodipine/valsartan/HCTZ administration, the GMRs (90%CI) of AUC0‐∞ and Cmax were 1.09 (1.04–1.15) and 1.11 (1.05–1.08) for amlodipine, 1.14 (0.99–1.31) and 1.12 (0.98–1.29) for valsartan, and 1.09 (1.02–1.16) and 0.86 (0.79–0.93) for HCTZ, respectively. Considering the sample size and pharmacokinetic variability associated with analytes, these study results indicate that food effect is minimal or none when fixed dose combination tablets are administered with food. In conclusion, both fixed dose combination tablets can be administered without regards to meals.


British Journal of Clinical Pharmacology | 2018

Effect of the angiotensin receptor–neprilysin inhibitor sacubitril/valsartan on the pharmacokinetics and pharmacodynamics of a single dose of furosemide

Surya Ayalasomayajula; Uwe Schuehly; Parasar Pal; Fabian Chen; Wei Zhou; Gangadhar Sunkara; Thomas Langenickel

Aims Sacubitril/valsartan is indicated for the treatment of heart failure and reduced ejection fraction (HFrEF). Furosemide, a loop diuretic commonly used for the treatment of HFrEF, may be coadministered with sacubitril/valsartan in clinical practice. The effect of sacubitril/valsartan on the pharmacokinetics and pharmacodynamics of furosemide was evaluated in this open label, two‐period, single‐sequence study in healthy subjects. Methods All subjects (n = 28) received 40 mg oral single‐dose furosemide during period 1, followed by a washout of 2 days. In period 2, sacubitril/valsartan 200 mg (97/103 mg) was administered twice daily for 5 days and a single dose of 40 mg furosemide was coadministered on day 6. Serial plasma and urine samples were collected to determine the pharmacokinetics of furosemide and sacubitril/valsartan and the pharmacodynamics of furosemide. The point estimates and the associated 90% confidence intervals for pharmacokinetic parameters were evaluated. Results Coadministration of furosemide with sacubitril/valsartan decreased the maximum observed plasma concentration (Cmax) [estimated geometric mean ratio (90% confidence interval): 0.50 (0.44, 0.56)], area under the plasma concentration–time curve (AUC) from time 0 to infinity [0.72 (0.67, 0.77)] and 24‐h urinary excretion of furosemide [0.74 (0.69, 0.79)]. When coadministered with sacubitril/valsartan, 0–4‐h, 4–8‐h and 0–24‐h diuresis in response to furosemide was reduced by ~7%, 21% and 0.2%, respectively, while natriuresis was reduced by ~ 28.5%, 7% and 15%, respectively. Post hoc analysis of the pivotal phase III Prospective comparison of ARNI with ACEI to Determine Impact on Global Mortality and morbidity in Heart Failure trial (PARADIGM‐HF) indicated that the median furosemide dose was similar at baseline and at the end of the study in the sacubitril/valsartan group. Conclusions Sacubitril/valsartan reduced plasma Cmax and AUC and 24‐h urinary excretion of furosemide, while not significantly affecting its pharmacodynamic effects in healthy subjects.

Collaboration


Dive into the Surya Ayalasomayajula's collaboration.

Researchain Logo
Decentralizing Knowledge