Skip to main content

Advertisement

Log in

Bayesian Two-Stage Biomarker-Based Adaptive Design for Targeted Therapy Development

  • Published:
Statistics in Biosciences Aims and scope Submit manuscript

Abstract

We propose a Bayesian two-stage biomarker-based adaptive randomization (AR) design for the development of targeted agents. The design has three main goals: (1) to test the treatment efficacy, (2) to identify prognostic and predictive markers for the targeted agents, and (3) to provide better treatment for patients enrolled in the trial. To treat patients better, both stages are guided by the Bayesian AR based on the individual patient’s biomarker profiles. The AR in the first stage is based on a known marker. A Go/No-Go decision can be made in the first stage by testing the overall treatment effects. If a Go decision is made at the end of the first stage, a two-step Bayesian lasso strategy will be implemented to select additional prognostic or predictive biomarkers to refine the AR in the second stage. We use simulations to demonstrate the good operating characteristics of the design, including the control of per-comparison type I and type II errors, high probability in selecting important markers, and treating more patients with more effective treatments. Bayesian adaptive designs allow for continuous learning. The designs are particularly suitable for the development of multiple targeted agents in the quest of personalized medicine. By estimating treatment effects and identifying relevant biomarkers, the information acquired from the interim data can be used to guide the choice of treatment for each individual patient enrolled in the trial in real time to achieve a better outcome. The design is being implemented in the BATTLE-2 trial in lung cancer at the MD Anderson Cancer Center.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. DiMasi JA, Hansen RW, Grabowski HG (2003) The price of innovation: new estimates of drug development costs. J Health Econ 22(2):151–185

    Article  Google Scholar 

  2. Herper M (2012) The truly staggering cost of inventing new drugs. Forbes Website. http://www.forbes.com/sites/matthewherper/2012/02/10/the-truly-staggering-cost-of-inventing-new-drugs/

  3. CSDD Outlook (2009) Backgrounder: a methodology for counting cost for pharmaceutical R&D. Tufts Center for the study of Drug Development, Boston

  4. Kola I, Landis J (2004) Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov 3:711–716

    Article  Google Scholar 

  5. Spear BB, Heath-Chiozzi M, Huff J (2001) Clinical application of pharmacogenetics. Trends Mol Med 7:201–204

    Article  Google Scholar 

  6. Simon R (2010) Clinical trial designs for evaluating the medical utility of prognostic and predictive biomarkers in oncology. Pers Med 7(1):33–47

    Article  Google Scholar 

  7. Mandrekar SJ, Sargent DJ (2009) Clinical trial designs for predictive biomarker validation: theoretical considerations and practical challenges. J Clin Oncol 27(24):4027–4034

    Article  Google Scholar 

  8. Mandrekar SJ, Sargent DJ (2001) Design of clinical trials for biomarker research in oncology. Clin Invest 1(12):1629–1636

    Google Scholar 

  9. Wang SJ (2007) Biomarker as a classifier in pharmacogenomics clinical trials: a tribute to 30th anniversary of PSI. Pharm Stat 6(4):283–296

    Article  Google Scholar 

  10. Scher HI, Nasso SF, Rubin EH, Simon R (2011) Adaptive clinical trial designs for simultaneous testing of matched diagnostics and therapeutics. Clin Cancer Res 17(21):6634–6640

    Article  Google Scholar 

  11. Goozner M (2012) Drug approvals 2011: focus on companion diagnostics. J Natl Cancer Inst 104(2):84–86

    Article  Google Scholar 

  12. Poste G, Carbone DP, Parkinson DR, Verweij J, Hewitt SM, Jessup JM (2012) Leveling the playing field: bringing development of biomarkers and molecular diagnostics up to the standards for drug development. Clin Cancer Res 18(6):1515–1523

    Article  Google Scholar 

  13. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144(5):646–674

    Article  Google Scholar 

  14. Reck M, Gatzemeier U (2004) Chemotherapy in stage-IV NSCLC. Lung Cancer 45:s217–222

    Article  Google Scholar 

  15. Schiller JH, Harrington D, Belani CP, Langer C, Sandler A, Krook J, Zhu J, Johnson DH for the Eastern Cooperative Oncology Group (2002) Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 346:92–98

    Article  Google Scholar 

  16. Silvestri G, Rivera M (2005) Targeted therapy for the treatment of advanced non-small cell lung cancer: a review of the epidermal growth factor receptor antagonists. Chest 183:29–42

    Google Scholar 

  17. McClellan M, Benner J, Schilsky R et al (2011) An accelerated pathway for targeted cancer therapies. Nat Rev Drug Discov 10:79–80

    Article  Google Scholar 

  18. Chabner BA (2011) Early accelerated approval for highly targeted cancer drugs. N Engl J Med 364(12):1087–1089

    Article  Google Scholar 

  19. Bates SE, Amiri-Kordestani L, Giaccone G (2012) Drug development: portals of discovery. Clin Cancer Res 18(1):23–32

    Article  Google Scholar 

  20. Sharma MR, Schilsky RL (2012) Role of randomized phase III trials in an era of effective targeted therapies. Nat Rev Clin Oncol 9(4):208–214

    Article  Google Scholar 

  21. Rubin EH, Gilliland DG (2012) Drug development and clinical trials–the path to an approved cancer drug. Nat Rev Clin Oncol 9(4):215–222

    Article  Google Scholar 

  22. Zhou X, Liu SY, Kim ES, Herbst RS, Lee JJ (2008) Bayesian adaptive design for targeted therapy development in lung cancer–a step toward personalized medicine. Clin Trials 5:181–193

    Article  Google Scholar 

  23. Kim ES, Herbst RS, Wistuba II, Lee JJ, Blumenschein GR, Tsao A, Stewart DJ, Hicks ME, Erasmus J, Gupta S, Alden CM, Liu S, Tang X, Khuri FR, Tran HT, Johnson BE, Heymach JV, Mao L, Fossella F, Kies MS, Papadimitrakopoulou V, Davis SE, Lippman SM, Hong WK (2011) The BATTLE trial: Personalizing therapy for lung cancer. Cancer Discov 1(1):44–53

    Article  Google Scholar 

  24. Berry DA (2005) Introduction to Bayesian methods III: use and interpretation of Bayesian tools in design and analysis. Clin Trials 2:295–300; discussion 301–304, 364–378

  25. Berry DA (2006) A guide to drug discovery: Bayesian clinical trials. Nat Rev Drug Discov 5:27–36

    Article  Google Scholar 

  26. Berry DA (2011) Adaptive clinical trials in oncology. Nat Rev Clin Oncol 9(4):199–207

    Article  Google Scholar 

  27. Hu F, Rosenberger WF (2003) Optimality, variability, power: evaluating response-adaptive randomization procedures for treatment comparisons. J Am Stat Assoc 98:671–678

    Article  MathSciNet  MATH  Google Scholar 

  28. Hu F, Rosenberger WF (2006) The theory of response-adaptive randomization in clinical trials. Wiley, Hoboken

    Book  MATH  Google Scholar 

  29. Thall PF (2002) Ethical issues in oncology biostatistics. Stat Methods Med Res 11:429–448

    Article  MATH  Google Scholar 

  30. Berry DA (2004) Bayesian statistics and the efficiency and ethics of clinical trials. Stat Sci 19:175–187

  31. Korn EL, Freidlin B (2010) Outcome-adaptive randomization: is it useful? J Clin Oncol 21:100–120

    Google Scholar 

  32. Berry DA (2010) Adaptive clinical trials: the promise and the caution. J Clin Oncol 21:606–609

    Google Scholar 

  33. Lee JJ, Chen N, Yin G (2012) Worth adapting? Revisiting the usefulness of outcome-adaptive randomization. Clin Cancer Res 18(17):4498–4507

    Article  Google Scholar 

  34. Barker AD et al (2009) I-SPY 2: an adaptive breast cancer trial design in the setting of neoadjuvant chemotherapy. Clin Pharm Ther 86:97–100

    Article  Google Scholar 

  35. Berry SM, Carlin BP, Lee JJ, Müller P (2010) Bayesian adaptive methods for clinical trials. Chapman & Hall, Boca Raton

    Book  MATH  Google Scholar 

  36. Lee JJ, Gu X, Liu S (2010) Bayesian adaptive randomization designs for targeted agent development. Clin Trials 7(5):584–596

    Article  Google Scholar 

  37. Eickhoff JC, Kim K, Beach J, Kolesar JM, Gee JR (2010) A Bayesian adaptive design with biomarkers for targeted therapies. Clin Trials 7:546–556

    Article  Google Scholar 

  38. Lara PN, Natale R, Crowley J et al (2009) Phase III trial of irinotecan/cisplatin Compared with etoposide/cisplatin in extensive-stage small-cell lung cancer: clinical and pharmacogenomic results from SWOG S0124. J Clin Oncol 27:2530–2535

    Article  Google Scholar 

  39. Park T, Casella G (2008) The Bayesian lasso. J Am Stat Assoc 103(482):681–686

    Article  MathSciNet  MATH  Google Scholar 

  40. Zou H (2006) The adaptive lasso and its oracle properties. J Am Stat Assoc 101(476):1418–1429

    Article  MathSciNet  MATH  Google Scholar 

  41. Kyung M, Gill J, Ghosh M (2010) Penalized regression, standard errors, and Bayesian lassos. Bayesian Anal 5:369–412

    Article  MathSciNet  MATH  Google Scholar 

  42. Meier L, van de Geer S, Buhlmann P (2008) The group lasso for logistic regression. J R Stat Soc B 70:53–71

    Article  MathSciNet  MATH  Google Scholar 

  43. Chipman H (1996) Bayesian variable selection with related predictors. Can J Stat 24:17–36

    Article  MathSciNet  MATH  Google Scholar 

  44. McCullagh P, Nelder J (1989) Generalized linear models. Chapman and Hall, Boca Raton

    Book  MATH  Google Scholar 

  45. Barbieri MM, Berger J (2004) Optimal predictive model selection. Ann Stat 32(3):870–897

    Article  MathSciNet  MATH  Google Scholar 

  46. Brannath W, Zuber E, Branson M, Bretz F, Gallo P, Posch M, Racine-Poon A (2009) Confirmatory adaptive designs with Bayesian decision tools for a targeted therapy in oncology. Stat Med 28(10):1445–1463

    Article  MathSciNet  Google Scholar 

  47. Biswas S, Liu DD, Lee JJ, Berry DA (2009) Bayesian clinical trials at the University of Texas MD Anderson Cancer Center. Clin Trials 6(3):205–216

    Article  Google Scholar 

  48. Lee JJ, Chu CT (2012) Bayesian clinical trials in action. Stat Med 31(25):2955–2972

    Article  MathSciNet  Google Scholar 

  49. Wang S-J, Li M-C (2013) Impacts of predictive genomic classifier performance on subpopulation-specific treatment effects assessment. Stat Biosci. doi:10.1007/s12561-013-9092-y

Download references

Acknowledgments

The authors thank Ms. LeeAnn Chastain for editorial assistance. The work was supported in part by grants CA016672 and CA155196 from the National Cancer Institute. The clinical trial was supported in part by Merck Research Laboratories and Bayer HealthCare. We also would like to thank two anonymous reviewers, the associate editor, and the editor for their thorough review and constructive critiques. Our manuscript has been improved by providing answers in addressing these critics.

Conflict of interest

The authors did not have any conflict of interest related to this work to disclose.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to J. Jack Lee.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (docx 48 KB)

Appendices

Appendix 1

The prior for \(\eta _k \) in (8) can be obtained by integrating out \(\tau _k^2 \):

$$\begin{aligned}&\int _{0}^{\infty }{\left( {\frac{{\tau _{k}^{2} }}{{2\pi }}} \right) ^{{\frac{{m_{k} }}{2}}} } \exp \left[ { - \frac{{\left\| {\eta _{k} } \right\| ^{2} }}{2}\tau _{k}^{2} } \right] \frac{{\left( {\frac{{\lambda _{k}^{2}}}{2}} \right) ^{{\frac{{m_{k} + 1}}{2}}} }}{{\varGamma \left( {\frac{{m_{k} + 1}}{2}} \right) }}(\tau _{k}^{2} )^{{ - \frac{{m_{k} + 1}}{2} - 1}} \exp \left[ { - \frac{{\lambda _{k}^{2} }}{{2\tau _{k}^{2} }}} \right] \mathrm{d}\tau _{k}^{2}\\&\quad = \int _{0}^{\infty } {\left( \! {\frac{1}{{2\pi }}} \!\right) ^{{\frac{{m_{k} }}{2}}} } \frac{{\left( \! {\frac{{\lambda _{k}^{2} }}{2}} \!\right) ^{{\frac{{m_{k} + 1}}{2}}} }}{{\varGamma \left( \! {\frac{{m_{k} + 1}}{2}} \!\right) }}\left( \! {\frac{1}{{(\tau _{k}^{2} )^{3} }}} \!\right) ^{{\frac{1}{2}}} \exp \left[ { - \frac{{\left\| {\eta _{k} } \right\| ^{2} \left( \! {\tau _{k}^{2} - \frac{{\lambda _{k} }}{{\left\| {\eta _{k} } \right\| }}} \!\right) ^{2} }}{{2\tau _{k}^{2} }} - \lambda _{k} \left\| {\eta _{k} } \right\| } \!\right] \mathrm{d}\tau _{k}^{2}\\&\quad = \int _{0}^{\infty } {\!\left( \! {\frac{1}{{2\pi }}} \!\right) ^{{\frac{{m_{k} }}{2}}} } \frac{{\left( \! {\frac{{\lambda _{k}^{2} }}{2}} \!\right) ^{{\frac{{m_{k} + 1}}{2}}} }}{{\varGamma \left( \! {\frac{{m_{k} + 1}}{2}} \!\right) }}\left( \! {\frac{{\lambda _{k}^{2} }}{{(\tau _{k}^{2} )^{3} }}} \!\right) ^{{\frac{1}{2}}} \!\exp \left[ \! { - \frac{{\lambda _{k}^{2} \left( \! {\tau _{k}^{2} - \frac{{\lambda _{k} }}{{\left\| {\eta _{k} } \right\| }}} \!\right) ^{2} }}{{2\frac{{\lambda _{k}^{2} }}{{\left\| {\eta _{k} } \right\| ^{2} }}\tau _{k}^{2} }}} \right] \exp [ - \lambda _{k} \left\| {\eta _{k} } \right\| ]\mathrm{d}\tau _{k}^{2}\\&\quad = \left( {\frac{1}{{2\pi }}} \right) ^{{\frac{{m_{k} - 1}}{2}}} \frac{{\left( {\frac{1}{2}} \right) ^{{\frac{{m_{k} + 1}}{2}}} }}{{\varGamma \left( {\frac{{m_{k} + 1}}{2}} \right) }}\lambda _{k}^{{m_{k} }} \exp [ - \lambda _{k} \left\| {\eta _{k} } \right\| ]\int _{0}^{\infty } {\left( {\frac{{\lambda _{k}^{2} }}{{2\pi (\tau _{k}^{2} )^{3} }}} \right) ^{{\frac{1}{2}}} }\\ {}&\qquad \ \times \exp \left[ { - \frac{{\lambda _{k}^{2} \left( {\tau _{k}^{2} - \frac{{\lambda _{k} }}{{\left\| {\eta _{k} } \right\| }}} \right) ^{2} }}{{2\frac{{\lambda _{k}^{2} }}{{\left\| {\eta _{k} } \right\| ^{2} }}\tau _{k}^{2} }}} \right] \mathrm{d}\tau _{k}^{2} \end{aligned}$$

The pdf of inverted Gaussian distribution is

$$\begin{aligned} \left( {\frac{\gamma }{2\pi x^3}}\right) ^{\frac{1}{2}}\exp \left[ {-\frac{\gamma ( {x-\mu })^2}{2\mu x}} \right] . \end{aligned}$$

When letting \(\tau _k^2 =x\), \(\frac{\lambda _k }{\left\| {\eta _k } \right\| }=\mu \), \(\lambda _k^2 =\gamma \), and integrating out \(\tau _k^2 \), we get the prior in (8).

Appendix 2

We used the same true scenario in Table 2 to simulate true patient response. However, instead of randomizing patients based on individual biomarker information (BATTLE-2 design strategy), patients were randomized based on biomarker group information (BATTLE-1 design strategy). Following BATTLE-1 design strategy, 3 biomarkers with decreasing priorities were used to group patients into 4 groups. A patient will be in group 1 if the first marker (highest priority) is positive; otherwise, the patient will be in group 2 if the second marker is positive; otherwise, the patient will be in group 3 if the third marker is positive; and the patient will be in group 4 if all 3 biomarkers are negative.

For the null case with the BATTLE-1 design strategy, the 3 markers used to group patients were marker 3, 4, and 5. Three different alternative cases were simulated using BATTLE-1 design strategy to represent 3 different situations. In Alternative 1 case, we assumed that the 3 true predictive markers were correctly identified, and marker 3, 4 and 5 were used to group patients. Unless specified explicitly, we always assumed that the marker with lower index has higher priority. In Alternative 2 case, we used marker 3, 6, and 7 to group patients, which means that one true predictive marker has been assigned the highest priority by chance. In Alternative 3 case, we used 3 randomly chosen markers to group patients, and this is equivalent to grouping patients with no credible marker information. The same hypothesis testings for the Go/No-Go decisions as in the simulation for BATTLE-2 design were conducted at the end of stage 1, and the same interim futility analyses were implemented. To make the results comparable to BATTLE-2 design, KRAS mutation and prior erlotinib history, in additional to the marker group information, were always included in the adaptive randomization. The same models with KRAS mutation and prior erlotinib history in (1) and (2) were used in hypothesis testing, and the marker group information was only used in adaptive randomization. Therefore, the differences in simulation results of BATTLE-1 and 2 design strategies were due to differences in randomization strategies and the added variable selection in BATTLE-2:

  1. (1)

    BATTLE-1 design strategy: Markers were used to group patients, and group information was used in adaptive randomization in BATTLE-1 design;

  2. (2)

    BATTLE-2 design strategy: Individual biomarkers were subjected to variable selection and, upon being selected, markers were directly used in adaptive randomization in BATTLE-2 design.

For comparison purpose, we simulated the clinical trials by following the design strategy of BATTLE-1 trial, and the results for hypothesis testing at the end of stage 1 are shown in Supplemental Table 1. The patient responses were simulated from the same true scenarios in Table 2. The same decision criteria and modeling approaches of BATTLE-2 design were also used for BATTLE-1 simulation. The error rates were controlled at the same rate of 10 % for each experimental treatment. Therefore, the differences in operating characteristics between BATTLE-1 and BATTLE-2 simulations can be attributed to the different strategies of incorporating biomarker information in adaptive randomization. The simulation results for Alternative 1 case in Supplemental Table 1 show that, in the best scenario where the predictive markers were correctly identified before the start of the clinical trial, BATTLE-1 design has higher power on the treatment (arm 2) associated with the marker having the highest priority, and has lower power on the remaining 2 experimental treatments (arm 3 and 4). In the worst scenario where none of the true predictive markers were correctly identified before the start of the trial (Alternative 3 case), BATTLE-2 trial design has higher power on all experimental treatments. In practice, Alternative 3 case is more likely to happen than Alternative 1 case. Because the purpose of clinical trial is to study experimental treatment and the biomarkers associated the treatment. It is unlikely that we are going to have perfect biomarker information at the beginning of clinical trial.

The patient allocation results for BATTLE-1 design strategy are shown in Supplemental Table 2. As compared with the results in Table 5 for BATTLE-2 design, the predictive marker with the highest priority can allocate more patients to the beneficial treatment arm in the most optimal situation (Alternative 1 and 2 cases) where the predictive marker has been correctly identified before the trial. In all other cases, the BATTLE-1 design strategy failed to learn from ongoing trial to treat patients better.

If credible informative biomarkers are available, using pre-selected biomarkers will be more powerful [49]. Our simulation comparison with BATTLE-1 design strategy also demonstrated that. However, in the exploratory stage of study when no credible biomarker information is available, our simulations showed that combining biomarkers into biomarker groups may lead to higher error rate in biomarker selection.

We also simulated the first stage of the trial using an equal randomization. The results are shown in the Supplemental Table 3. Under the null hypothesis, when the overall error rate for the trial is still controlled at 20 %, the average error rate for each experimental treatment arm is about 1–2 % lower (0.077–0.091 under ER and 0.088–0.100 for AR). When the alternative hypothesis is true, the increase in average power for each experimental treatment arm can be as high as 15 % (0.97 to 0.99 for ER and 0.82 to 0.95 for AR). However, the increase of the overall power of the trial is only 2 % (0.999 for ER and 0.978 for AR). With AR and early stopping rule, it can result in a sample size saving of 60 patients under the null hypothesis. AR also resulted in more patients to be treated with more effective treatments.

Appendix 3

The likelihood function of logistic model can be expressed as:

$$\begin{aligned} \mathop \prod \limits _{i=1}^n \left[ {\left( {\frac{\exp ( {X_i \theta })}{1+\exp ({X_i \theta })}}\right) ^{y_i } \left( {\frac{1}{1+\exp ( {X_i \theta })}}\right) ^{1-y_i }} \right] =\frac{\exp ( {\mathop \sum \nolimits _{i\in \Omega _R } X_i \theta })}{( {1+\exp ( {X_i \theta })})^n}, \end{aligned}$$

where \(i\) is the index for individual patient, \(n\) is the total number of patients, \({{\varvec{\Omega }}}_R\) is the set of index for all responders, \(X_i \) is a generic representation of covariate, and \(\theta \) is a generic representation of model parameter. Without loss of generality, we assume that both \(X_i\) and \(\theta \) are scales.

For Bayesian adaptive lasso, the prior for \(\theta \) given \(\lambda \) is

$$\begin{aligned} \pi (\theta \vert \lambda )\propto \text{ exp }\left( -\lambda \frac{\vert \theta \vert }{\hat{\theta }}\right) , \end{aligned}$$

where \(\hat{\theta }\) is a square root n consistent initial estimate of \(\theta \). The square of \(\lambda \) follows a gamma distribution:

$$\begin{aligned} \pi ( {\lambda ^2})=\frac{\lambda ^{2(a-1)}e^{-\frac{\lambda ^2}{b}}}{b^a\varGamma (a)}, \end{aligned}$$

where \(a\) and \(b\) are hyper parameters.

The conditional posterior distributions of \(\theta \) and \(\lambda \) are

$$\begin{aligned} \theta \vert \lambda \propto \frac{\exp ( {\mathop \sum \nolimits _{i\in {{\varvec{\Omega }}}_R } X_i \theta })}{( {1+\exp ( {X_i \theta })})^n}{\cdot }\text{ exp } \left( {-\lambda \frac{\left| \theta \right| }{\hat{\theta }}}\right) \end{aligned}$$
$$\begin{aligned} \lambda \vert \theta \propto \lambda ^{2( {a-1})}e^{-\frac{\lambda ^2}{b}}{\cdot }\exp \left( {-\lambda \frac{\left| \theta \right| }{\hat{\theta }}}\right) . \end{aligned}$$

It is easy to show that all above conditional distributions are log-concave. Therefore, the adaptive rejection sampling implemented in OpenBugs can be conveniently utilized to generate the posterior distribution of model parameters for Bayesian adaptive lasso.

Appendix 4

For group parameter \({\varvec{\eta }}\) in Bayesian adaptive group lasso, the prior distribution is

$$\begin{aligned} \pi (\eta \mid \lambda )\propto \text{ exp }(-\lambda \vert \left| \eta \right| \vert /\vert |{\hat{{\varvec{\eta }}}}|\vert ), \end{aligned}$$

where \(\hat{{\varvec{\eta }}}\) is an initial estimate of \(\eta \). In our simulation, we used the gamma mixture of normals in Eq. (9). After \(\tau _k \) in Eq. (9) is integrated out, Appendix 1 shows that these two formulations are equivalent while conditional on \(\lambda \).

The same gamma prior distribution is assumed for the square of \(\lambda \):

$$\begin{aligned} \pi ( {\lambda ^2})=\frac{\lambda ^{2(a-1)}e^{-\frac{\lambda ^2}{b}}}{b^a \varGamma (a)} \end{aligned}$$

The likelihood function of logistic model can be expressed as:

$$\begin{aligned} \mathop \prod \limits _{i=1}^n \left[ {\left( {\frac{\exp ( {X_i^\mathrm{T} \eta })}{1+\exp ( {X_i^\mathrm{T} \eta })}}\right) ^{y_i } \left( {\frac{1}{1+\exp ( {X_i^\mathrm{T} \eta })}}\right) ^{1-y_i }} \right] =\frac{\exp ( {\mathop \sum \nolimits _{i\in {{\varvec{\Omega }}}_R } X_i^\mathrm{T} \eta })}{( {1+\exp ({X_i^\mathrm{T} \eta })})^n}. \end{aligned}$$

Both \(X_i \) and \(\eta \) are column vectors. Without loss of generality, we assume that there is only one group of variables in the model. Therefore, the posterior distribution of model parameters can be sampled from

$$\begin{aligned} \begin{array}{l} \eta ,\tau ,\lambda \vert X,Y \\ \propto \frac{\exp ( {\mathop \sum \nolimits _{i\in {{\varvec{\Omega }}}_R } X_i^\mathrm{T} \eta })}{( {1+\exp ( {X_i^\mathrm{T} \eta })})^n}\tau ^m\text{ exp }\left[ {-\frac{\left\| \eta \right\| ^2}{2}\tau ^2} \right] \lambda ^{m+1}\tau ^{-m-3}\text{ exp }\left[ {-\frac{\lambda ^2}{2 \Vert {\hat{{\varvec{\eta }}}}\Vert ^2\tau ^2}} \right] \lambda ^{2(a-1)}\exp \left[ {-\frac{\lambda ^2}{b}} \right] , \\ \end{array} \end{aligned}$$

where Y is a vector of observed response outcomes, \(m\) is the dimension of \(\eta \), and \(\left\| \eta \right\| ^2=\eta _1^2 +\cdots +\eta _m^2 \). The full conditional distributions are

$$\begin{aligned} \eta \vert \tau ,\lambda ,X,Y\propto \frac{\exp ( {\mathop \sum \nolimits _{i\in {{\varvec{\Omega }}}_R } X_i^\mathrm{T} \eta })}{( {1+\exp ( {X_i^\mathrm{T} \eta })})^n}\text{ exp }\left[ {-\frac{\left\| \eta \right\| ^2}{2}\tau ^2} \right] \end{aligned}$$
$$\begin{aligned} \tau \vert \eta ,\lambda ,X,Y\propto \tau ^{-3}\text{ exp }\left[ {-\frac{\lambda ^2}{2 \Vert {\hat{{\varvec{\eta }}}} \Vert ^2\tau ^2}} \right] \text{ exp }\left[ {-\frac{\left\| \eta \right\| ^2}{2}\tau ^2} \right] \end{aligned}$$
$$\begin{aligned} \lambda \vert \eta ,\tau ,X,Y\propto \lambda ^{2a+m-1}\text{ exp }\left[ {-\lambda ^2\left( {\frac{1}{b}+\frac{1}{2 \Vert {\hat{{\varvec{\eta }}}} \Vert ^2\tau ^2}}\right) } \right] . \end{aligned}$$

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gu, X., Chen, N., Wei, C. et al. Bayesian Two-Stage Biomarker-Based Adaptive Design for Targeted Therapy Development. Stat Biosci 8, 99–128 (2016). https://doi.org/10.1007/s12561-014-9124-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12561-014-9124-2

Keywords

Navigation