Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1206 word(s) 1206 2021-08-09 12:46:41

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Van Doorn, L. Scalp Cooling. Encyclopedia. Available online: https://encyclopedia.pub/entry/13425 (accessed on 18 April 2024).
Van Doorn L. Scalp Cooling. Encyclopedia. Available at: https://encyclopedia.pub/entry/13425. Accessed April 18, 2024.
Van Doorn, Leni. "Scalp Cooling" Encyclopedia, https://encyclopedia.pub/entry/13425 (accessed April 18, 2024).
Van Doorn, L. (2021, August 20). Scalp Cooling. In Encyclopedia. https://encyclopedia.pub/entry/13425
Van Doorn, Leni. "Scalp Cooling." Encyclopedia. Web. 20 August, 2021.
Scalp Cooling
Edit

Chemotherapy-induced alopecia (CIA), a side effect with high impact, can be prevented by cooling the scalp during the administration of some cytotoxic drugs. Scalp cooling is a well-known method to try to prevent CIA during the administration of cytotoxic drugs for solid tumors. Using scalp cooling, liquid refrigerant is pumped as coolant through a cooling cap that is placed on the head of the patient. In general, scalp cooling is started 20–45 min prior to, during, and up to 20–150 min after the chemotherapy infusion.

chemotherapy-induced alopecia scalp cooling paclitaxel clearance

1. Introduction

Chemotherapy-induced alopecia (CIA) is a commonly feared side effect of systemic anti-cancer treatment.[1] It can affect a patient’s quality of life dramatically and is one of the most distressing and adverse aspects of anti-cancer treatment, particularly for women.[2] Scalp cooling is a well-known method to try to prevent CIA during the administration of cytotoxic drugs for solid tumors.[3][4] Scalp cooling results in a locally decreased blood flow due to vasoconstriction, resulting in a lower chemotherapy concentration at the root of the hair follicles and thereby, hopefully, in hair preservation.[5] The pharmacokinetics and pharmacodynamics of several drugs are influenced by body temperature.[6] Deep scalp cooling (4 °C), lasting for 20–45 min before, continued during and lasting for up to 150 min after chemotherapy infusion, may potentially lead to a temperature reduction of the whole body. This drop in body temperature may lead to alterations in pharmacokinetics.[6] This is of clinical relevance as changes in pharmacokinetics may lead to under- or over-exposure to the drug of interest. In a previous study, a physiologically based pharmacokinetic model (PBPK) of doxorubicin was modified to include a scalp skin compartment. The results of the model showed that maximum and average concentrations of doxorubicin in the scalp skin compartment were reduced by a factor of 3.6 and 1.6, respectively, during scalp cooling. These effects were due to reduced tissue perfusion and can positively influence the survival of hair follicles. However, mass transfer characteristics were not considered.[7][8]
At present, there are no data available regarding the effects of scalp cooling on the pharmacokinetics of the cytotoxic drugs that are infused.
The severity of CIA, but also the success rate of scalp cooling, depends on the type of anti-cancer treatment used, its dose, method of administration and schedule of treatment.[9][10] Scalp cooling in patients treated with taxane-based chemotherapy such as paclitaxel, a widely used antineoplastic agent for the treatment of several cancers (e.g., breast, ovarian and esophageal cancer),[11][12][13] led to hair conservation in more than 50%, of patients, depending on the dose, compared with those who received no scalp cooling.[14] Scalp cooling is therefore offered as a part of standard treatment.

2. Effect of Scalp Cooling on the Pharmacokinetics of Paclitaxel

Blood samples for the pharmacological analyses were collected during one of the paclitaxel administrations, not necessarily the first administration, at four predefined time points: pre-dose, 55 (5 min prior to the end of the paclitaxel infusion), 90 and 360 min after the start of the paclitaxel infusion.[15] In patients where paclitaxel was administered in a standard stepwise increase in infusion rates because of hypersensitivity during previously administered paclitaxel treatment, this was pre-dose, 85 (5 min prior to the end of the paclitaxel infusion), 120 and 360 min after the start of paclitaxel infusion. The samples were collected by venipuncture or cannula in 4.5 mL lithium heparin blood collection tubes and processed within 10 min by centrifugation for 10 min at 2500–3000× g at 4°C. Plasma was transferred into polypropylene tubes (1.8 mL Nunc Cryotube vials), which were stored at a temperature of minus 70 °C. Paclitaxel pharmacokinetics were measured in all plasma samples using a validated ultra-performance liquid chromatographic coupled to tandem mass spectrometry (UPLC-MS/MS) for precise quantification of paclitaxel plasma concentrations at the Laboratory of Translational Pharmacology of the Erasmus MC Rotterdam.[16] Non-linear mixed effects modeling was conducted using the software NONMEM.[17] A previously developed population pharmacokinetic model for paclitaxel,[14] with two compartments describing the disposition and linear elimination, was used as a starting point. However, the model was here expanded to a three-compartment model to fit the data. Scalp cooling was tested for its effect on the model PK parameters.
Although the decrease in body temperature during scalp cooling at each site measured was small, the decrease was statistically significant different during scalp cooling for the measurement in the ear and axilla. There was no significant difference in temperature until 3h after scalp cooling was discontinued. Finally, half of the patients developed some form of hair loss despite scalp cooling. However, this was not associated with paclitaxel clearance.
Mild hypothermia (body cooling to 32 to 34°C for 12h to 48h) can alter the pharmacokinetic parameters of several drugs.[6] Although the mechanism(s) behind changes in drug levels due to hypothermia has not been fully elucidated, impaired hepatic metabolism is likely, possibly via its effect on cytochrome P450 metabolism. In a study in healthy volunteers, for example, the clearance of midazolam as an index of CYP3A4/5 metabolism decreased by 11% for every degree Celsius decrease in a core temperature of 36.5°C.[18]
In our analysis, a population PK model consisting of a central compartment with two peripheral compartments connecting to it was used to obtain clearances and volumes of distribution of each subject. Unlike PBPK models, these compartments have no anatomic or physiological significance. However, they can still be used to investigate the influence of subject characteristics (i.e., scalp cooling) on the predicted subject PK parameters describing the whole-body drug disposition.[19]
We hypothesized that the decrease in body temperature as a result of scalp cooling may influence the pharmacokinetics of paclitaxel. Although a small decrease in body temperature was found after 50 min of scalp cooling, which did not fully return to baseline after 5h, the absolute decrease was limited: 60% of the women had a temperature drop of less than just 1°C. This drop in temperature may be too small to demonstrate a difference in clearance of paclitaxel.
Scalp cooling is usually a good option to prevent hair loss for anti-cancer drugs with a short half-life or a rapid systemic distribution, such as paclitaxel.[10] However, it is unclear why some patients still develop CIA despite scalp cooling during such chemotherapy administration. Half of the women were found to have some form of hair loss, of whom more than 40% developed grade 2 alopecia (although no one developed full baldness). This is somewhat higher than mentioned in previous studies, probably due to differences in definition of CIA between these studies.[10] We found no clear difference between the patients who developed CIA and those who did not. If any, the mean temperatures were slightly lower among those women without CIA compared to those with CIA. It is important to emphasize that CIA was also not due to differences in paclitaxel clearance. Further research is needed to identify possible explanations to better advise future patients about the chance of hair preservation with scalp cooling.

3. Conclusions

Scalp cooling concomitant with paclitaxel did not reduce nor increase the clearance of paclitaxel. Therefore, it is unlikely that scalp cooling influences paclitaxel efficacy.
Finally, despite scalp cooling, half of the patients in our study developed a form of hair loss. Importantly, neither an association with difference in paclitaxel clearance nor a change in hair loss was found. Further research is warranted to optimize hair preservation in patients treated with paclitaxel.

References

  1. Dunnill, C.J.; Al-Tameemi, W.; Collett, A.; Haslam, I.S.; Georgopoulos, N.T. A clinical and biological guide for understanding chemotherapy-induced alopecia and its prevention. Oncologist 2018, 23, 84–96.
  2. Van den Hurk, C.J.; Mols, F.; Vingerhoets, A.J.; Breed, W.P. Impact of alopecia and scalp cooling on the well-being of breast cancer patients. Psychooncology 2010, 19, 701–709.
  3. Van den Hurk, C.J.; van den Akker-van Marle, M.E.; Breed, W.P.; van de Poll-Franse, L.V.; Nortier, J.W.; Coebergh, J.W. Impact of scalp cooling on chemotherapy-induced alopecia, wig use and hair growth of patients with cancer. Eur. J. Oncol. Nurs. 2013, 17, 536–540.
  4. Massey, C.S. A multicentre study to determine the efficacy and patient acceptability of the paxman scalp cooler to prevent hair loss in patients receiving chemotherapy. Eur. J. Oncol. Nurs. 2004, 8, 121–130.
  5. Breed, W.; van den Hurk, C.J.G.; Peerboom, M. Presentation, impact and prevention of chemotherapy-induced hair loss. Expert Rev. Dermatol. 2011, 6, 109–125.
  6. Van den Broek, M.P.; Groenendaal, F.; Egberts, A.C.; Rademaker, C.M. Effects of hypothermia on pharmacokinetics and pharmacodynamics: A systematic review of preclinical and clinical studies. Clin. Pharm. 2010, 49, 277–294.
  7. Gustafson, D.L.; Rastatter, J.C.; Colombo, T.; Long, M.E. Doxorubicin pharmacokinetics: Macromolecule binding, metabolism, and excretion in the context of a physiologic model. J. Pharm. Sci. 2002, 91, 1488–1501.
  8. Janssen, F.E.M. Modelling Physiological and Biochemical Aspects of Scalp Cooling. Ph.D. Thesis, Technical University Eindhoven, Eindhoven, The Netherlands, 2007.
  9. Van den Hurk, C.J.; Peerbooms, M.; van de Poll-Franse, L.V.; Nortier, J.W.; Coebergh, J.W.; Breed, W.P. Scalp cooling for hair preservation and associated characteristics in 1411 chemotherapy patients—Results of the dutch scalp cooling registry. Acta Oncol. 2012, 51, 497–504.
  10. Komen, M.M.; Smorenburg, C.H.; van den Hurk, C.J.; Nortier, J.W. Factors influencing the effectiveness of scalp cooling in the prevention of chemotherapy-induced alopecia. Oncologist 2013, 18, 885–891.
  11. Stage, T.B.; Bergmann, T.K.; Kroetz, D.L. Clinical pharmacokinetics of paclitaxel monotherapy: An updated literature review. Clin. Pharm. 2018, 57, 7–19.
  12. Van Hagen, P.; Hulshof, M.C.; van Lanschot, J.J.; Steyerberg, E.W.; van Berge Henegouwen, M.I.; Wijnhoven, B.P.; Richel, D.J.; Nieuwenhuijzen, G.A.; Hospers, G.A.; Bonenkamp, J.J.; et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N. Engl. J. Med. 2012, 366, 2074–2084.
  13. Heijkoop, S.T.; Franckena, M.; Thomeer, M.G.; Boere, I.A.; Van Montfort, C.; Van Doorn, H.C. Neoadjuvant chemotherapy followed by radiotherapy and concurrent hyperthermia in patients with advanced-stage cervical cancer: A retrospective study. Int J. Hyperth. 2012, 28, 554–561.
  14. Nangia, J.; Wang, T.; Osborne, C.; Niravath, P.; Otte, K.; Papish, S.; Holmes, F.; Abraham, J.; Lacouture, M.; Courtright, J.; et al. Effect of a scalp cooling device on alopecia in women undergoing chemotherapy for breast cancer: The scalp randomized clinical trial. JAMA 2017, 317, 596–605.
  15. Henningsson, A.; Sparreboom, A.; Sandstrom, M.; Freijs, A.; Larsson, R.; Bergh, J.; Nygren, P.; Karlsson, M.O. Population pharmacokinetic modelling of unbound and total plasma concentrations of paclitaxel in cancer patients. Eur. J. Cancer 2003, 39, 1105–1114.
  16. De Graan, A.J.; Elens, L.; Sprowl, J.A.; Sparreboom, A.; Friberg, L.E.; van der Holt, B.; de Raaf, P.J.; de Bruijn, P.; Engels, F.K.; Eskens, F.A.; et al. Cyp3a4*22 genotype and systemic exposure affect paclitaxel-induced neurotoxicity. Clin. Cancer Res. 2013, 19, 3316–3324.
  17. Bauer, R.J. Nonmem tutorial part i: Description of commands and options, with simple examples of population analysis. CPT Pharmacomet. Syst. Pharm. 2019, 8, 525–537.
  18. Hostler, D.; Zhou, J.; Tortorici, M.A.; Bies, R.R.; Rittenberger, J.C.; Empey, P.E.; Kochanek, P.M.; Callaway, C.W.; Poloyac, S.M. Mild hypothermia alters midazolam pharmacokinetics in normal healthy volunteers. Drug Metab Dispos. 2010, 38, 781–788.
  19. Mould, D.R.; Upton, R.N. Basic concepts in population modeling, simulation, and model-based drug development. CPT Pharmacomet. Syst. Pharm. 2012, 1, e6.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 234
Revision: 1 time (View History)
Update Date: 20 Aug 2021
1000/1000