You are here: Home: BCU 1|2003: Nancy Davidson, MD

Nancy Davidson, MD

Professor of Oncology,
Breast Cancer Research Chair in Oncology,
The Johns Hopkins University School of Medicine

Director, Breast Cancer Research Program,
The Johns Hopkins Oncology Center

Chair, Breast Committee,
Eastern Cooperative Oncology Group

Edited comments by Dr Davidson

Postmastectomy radiation therapy in women with one to three positive nodes

I usually have my node-positive, post-mastectomy patients evaluated by a radiation oncologist to discuss the pros and cons of radiation. If they have four or more nodes, I recommend radiation pretty highly, and if they’re nodenegative I’m pretty much against it. In my experience, younger women with greater numbers of positive lymph nodes are more likely to opt for radiation.

Radiation therapy decisions are also often influenced by the type of reconstruction that a woman has had. Women with implant reconstruction are sometimes not quite as enthusiastic about radiation because of the potential deleterious cosmetic effects.

Nonprotocol use of AC-docetaxel

We participate in the NSABP B-30 trial, which involves AC followed by docetaxel as its standard arm. In a nontrial setting, I would frequently think about using the standard arm. For example, if I was discussing NSABP B-30 with a patient, when we come back to a discussion of standard therapy outside of the trial, we would talk about the standard arm of this trial.

I would tell her about our uncertainty with regard to the taxanes. Sometimes in a nonprotocol setting, we go in with the notion that the patient is going to take the AC, we’ll see how it is going and then she’ll come back and tell me how she feels about taking the taxane. I have not been a big fan of six cycles of TAC or FEC, but I know that many physicians are.

Docetaxel versus paclitaxel in the adjuvant setting

Some physicians are more interested in docetaxel than paclitaxel for several reasons. One is the enthusiasm about the preoperative docetaxel results from NSABP B-27. The second reason is that some people have looked at the BCIRG trial of TAC versus FAC as an endorsement of docetaxel.

I think that we’re doing an awful lot of early reporting. The TAC results are interesting, but I want to see more follow-up. I actually thought TAC would make a lot of headway in the community, but — at least where I live — it doesn’t seem to have made a big impact.

I'm most impressed that people are taking the subset analysis from that trial very seriously. I’ve had people call me, reluctant to use TAC in a patient with six positive lymph nodes, because in that trial the advantage was only seen in the women with one to three positive nodes. I'm impressed with how evidence-based many of the physicians that I have spoken to are in making therapeutic decisions.

 

Non-anthracycline containing regimens

We all feel reasonably confident that anthracycline-containing regimens are probably slightly better than non-anthracycline-containing regimens; however, I am still a fan of CMF in patients who have cardiotoxicity issues. If you look at the differences — for example a CAF versus CMF trial that we did through the Intergroup — the absolute difference in benefit was actually very small. Some patients may not find that worthwhile when considering the tradeoff in terms of the cardiotoxicity concerns.

Effect of HER2 and nodal status on choice of chemotherapy

I have not routinely used HER2 status to make chemotherapy decisions. I do tell patients that there is some belief that HER2 positivity might drive one to think harder about an anthracycline-containing regimen. This finding, however, isn’t true across all studies, and we know from our adjuvant trastuzumab trials that we’re not very good at measuring HER2 status.

There has been as much as a 25-30 percent discordance rate between local and central laboratory testing. This makes me very nervous about putting a lot of emphasis on a study if I’m not completely confident about the quality of the data.

If a patient had 15 positive nodes, I would probably think even harder about any regimen that involves six months of therapy and not so hard about four cycles of AC. I would also be thinking very hard about her endocrine therapy. With regard to adjuvant trastuzumab, I am a purist on this issue and a big believer in the randomized trials — I have not given any adjuvant trastuzumab outside the context of a clinical trial.

Ovarian suppression in ER-positive, premenopausal women

Many younger women are still menstruating after the completion of chemotherapy. Several years ago, I would only have discussed tamoxifen, but presently I do actually discuss the uncertainty about ovarian suppression strategies. I usually recommend tamoxifen, and if a patient feels strongly, sometimes she’ll also undergo ovarian suppression.

In higher-risk women, I would consider it more strongly. Based on a very small retrospective subset analysis, women with 10 or more positive lymph nodes were the ones who seemed to get a fair amount of benefit from the combined endocrine therapy. The caveat here is that even in our seemingly large trial that, that group is only 100 women — a very small subset to base a lot on.

Impact of HER2 status on choice of endocrine therapy

There is a belief that perhaps aromatase inhibitors are more effective than tamoxifen in ER-positive, postmenopausal women whose tumors overexpress HER2. This is based, in part, on Matt Ellis’ preoperative study. There is really no equivalent data in premenopausal women.

Richard Love did a trial in Vietnam of premenopausal women where the standard of care was no adjuvant therapy, and the experimental arm was oophorectomy and tamoxifen. He found that there was no impact of HER2 status on response to endocrine therapy. Combined endocrine therapy was effective regardless of HER2 status. I don’t think HER2 status should have any influence on the approach to adjuvant endocrine therapy.

If a premenopausal woman stops menstruating after the completion of chemotherapy, I would be oriented towards tamoxifen, but I would have a discussion about tamoxifen versus anastrozole. Many of my very sophisticated patients would want to talk about the impact of HER2 status in that setting, and I’ve had a couple who decided to go on anastrozole because of their belief that tamoxifen may not be as good in that subset of ER-positive, HER2-positive women.

Endocrine therapy in a woman with history of a deep vein thrombosis

In a postmenopausal woman who has had a deep vein thrombosis in the past several years, I would move to an aromatase inhibitor without thinking very hard about it. If she had stopped menstruating after chemotherapy, I would probably consider her postmenopausal.

It’s a little tougher in premenopausal women, and I would think more about an ovarian suppression strategy as my only strategy. Ovarian ablation or suppression as an alternative to tamoxifen in premenopausal patients was endorsed by the 2000 NIH consensus conference.

Intergroup Trial 0101

The design of this trial was CAF chemotherapy versus CAF chemotherapy followed by five years of goserelin versus CAF chemotherapy followed by five years of goserelin and tamoxifen. There is no impact on disease-free survival in the overall population with the addition of goserelin, but there is a trend to suggest that the younger patients may benefit.

Although it seemed like such a large clinical trial at the time it was initiated, 1,500 women doesn’t have the power to reveal a significant difference even in those younger women and even with all this follow-up.

We don’t have any new data over the last year on this question. We have a lot of re-examination of old data. My synopsis is that in ER-positive, premenopausal women, tamoxifen is a good drug. Ovarian suppression or ablation is also beneficial, but we are having a difficult time figuring out how to integrate them.

The one new trial that I’ve seen over the last year is the Austrian trial published in the last couple of months comparing CMF chemotherapy to ovarian suppression with tamoxifen in premenopausal estrogen receptorpositive women. They suggested that the outcome was slightly better with the combined endocrine therapy.

The difficulty with that trial is that the women who took chemotherapy didn’t take tamoxifen because it was not the standard of care when the trial was launched. Today we think of that as a pretty profound deficit with that study and related studies. So we need to come together to investigate this further. There is a trio of trials that we are trying to launch worldwide to look at issues of ovarian suppression in young women.

Combining LHRH agonists and aromatase inhibitors in premenopausal women

I’m very enthusiastic about the research strategy of looking at LHRH agonists with aromatase inhibitors. Extrapolating from the early data in postmenopausal breast cancer, which suggested that anastrozole may have superior efficacy compared to tamoxifen, this seems like a rational strategy to transfer to premenopausal women as well. The two issues are whether or not it is actually going to be efficacious, and what is the cost in terms of side effects. I wouldn’t utilize this strategy outside the context of a clinical trial.

Assessment of menopausal status in ER-positive patients and choice of endocrine therapy

In terms of determining whether a woman is pre- or postmenopausal, I usually just assess patients clinically, not by testing with blood work. If their menstrual periods go away, usually I’m already giving tamoxifen if the patient is ER-positive, so I don’t actually need to know her menopausal status to approach that. If we were routinely using anastrozole in postmenopausal women — and we are in that transition time right now — then we might have to work a little harder to make sure they truly are postmenopausal. The other issue is that women can become transiently postmenopausal and have recovery of ovarian function at a later date.

I approach premenopausal women with metastatic disease who become clinically menopausal after receiving chemotherapy as postmenopausal. I have been using first-line aromatase inhibitors in these women for several reasons. First, I’m impressed by the trial data comparing them to tamoxifen as first-line therapy. Second, many of those women have already been exposed to or are on tamoxifen at the time of their relapse.

I start with either letrozole or anastrozole, and I can’t tell you why sometimes I choose one over the other. I generally do not use exemestane as my first choice. If the person has a good response to their first aromatase inhibitor, I am hoping to capitalize on the work from Per Lonning suggesting that women who have been exposed to the nonsteroidal aromatase inhibitors can have a 20 percent clinical benefit with exemestane.

SOFT: Ovarian ablation with tamoxifen or an aromatase inhibitor

The adjuvant ovarian suppression trial that I am most enthusiastic about is SOFT — Suppression of Ovarian Function Trial. Premenopausal, ER-positive women who may or may not have received chemotherapy will be randomized to tamoxifen for five years, ovarian suppression/ablation plus tamoxifen, or ovarian suppression/ablation plus an aromatase inhibitor. This very interesting trial will help us address several issues. Does ovarian ablation or suppression add to tamoxifen? And if this is an important strategy, is it better to use tamoxifen or an aromatase inhibitor in those suppressed women?

This trial is an international collaboration, put together by the International Breast Cancer Study Group (IBCSG). The US cooperative groups have signed on to it, and it is winding its way through the approval process in the United States right now. I think it will be launched within the next year.

Other trials of aromatase inhibitors with ovarian suppression

There are two other studies of aromatase inhibitors with ovarian suppression. One is built on the premise — which is pretty popular in Europe — that since we know ovarian suppression is important, some investigators would be unenthusiastic about a trial that didn’t involve ovarian suppression. For those investigators, the trial would be ovarian suppression with tamoxifen or ovarian suppression with an aromatase inhibitor.

The other trial asks the question, “If you do ovarian suppression with either tamoxifen or an aromatase inhibitor, do you really need chemotherapy?” This trial randomizes to chemotherapy or not, plus endocrine therapy.

I believe that will be a tough concept to sell in the United States, but it may have some enthusiasts abroad. I personally think randomized trials that involve two very different treatments, chemotherapy or not, will be a little more difficult to conceptualize.

These trials were put together by the International Breast Cancer Study Group. They have been looked at by the US cooperative groups, and different groups will decide whether or not to endorse each trial. Subgroups may decide that they are not as enthusiastic about one design or another, and that they want to put all their effort into one. My personal preference is the SOFT trial, because I think it addresses the issues of interest to many US investigators.

Counseling postmenopausal women on adjuvant endocrine therapy: Tamoxifen versus aromatase inhibitors

In counseling women about adjuvant endocrine therapy, it’s a lot longer discussion now than in the past, because I feel obligated to go through the ATAC trial in some detail and talk with people about their preferences. Some women are pretty clear that they want anastrozole, and I am comfortable prescribing it to them. Obviously, if somebody has contraindications to tamoxifen, it’s a pretty easy decision.

Many patients know a lot about tamoxifen. They know that it has a long track record, and they’re pretty comfortable with that. However, it's always stunning to me as an oncologist how much bad press tamoxifen has received, for practically the least toxic drug I can prescribe. It amazes me how many people will go through six months of chemotherapy without batting an eyelash, yet come in very concerned about the long-term consequences of tamoxifen.

The majority of my patients are going on tamoxifen right now, but I think the sands are shifting. At the beginning, everybody sort of sat tight, but since the FDA approval, I’ve seen more women who are open to anastrozole by the time they come to see me. When I do use an aromatase inhibitor in the adjuvant setting, I only use anastrozole. I’m a purist on that. The one trial we have adjuvant data from utilized anastrozole, so I want to do it exactly as we did in that trial.

Use of bisphosophonates in the adjuvant setting

There is a trial in Austria randomizing premenopausal, ER-positive patients to various endocrine therapies with a second randomization to different schedules of zoledronate. Their long-term goal is not only impact on bone density, but also on breast cancer recurrence, based on all these conflicting results with clodronate.

In my practice, I usually watch bone mineral densities, and if they get down to a range I’m unhappy with, I use one of the oral bisphosphonates. I still use a lot of adjuvant tamoxifen, and for premenopausal women that is a pretty good bone drug. They may not need anything in addition until they come off of tamoxifen. I haven’t used a lot of adjuvant aromatase inhibitors, but I think this is where it might turn out to be an issue in the future.

Approach to chemotherapy in younger versus older women with metastatic disease

My philosophy in treating older versus younger women with chemotherapy is basically the same, but sometimes the patient choices are different. Many times in metastatic disease, we use all of the available therapies, so what we’re really deciding on is the order — what to start with. Many patients make that decision based on their personal values. I find many of my older patients are attracted to capecitabine because it is an oral agent. Some of my younger patients think of intravenous therapy as more aggressive, and they prefer that strategy. But, this perception is people’s gut reaction rather than being reality-based.

Capecitabine in the metastatic setting

I am a big fan of capecitabine. Maybe it comes from being a "hormonal-therapy person" preferring pills to begin with, because I use it a lot for salvage chemotherapy in women who’ve already had an anthracycline and taxane for metastatic disease. In oncology, we tend to remember our successes, but I have seen several very impressive responses with capecitabine in pretty dire circumstances. I have had women on it for a considerable period of time with relatively good quality of life. My personal best was somebody who was on capecitabine for several years.

Combination versus sequential therapy in the metastatic setting

ECOG-1193 trial compared doxorubicin followed by paclitaxel at disease progression versus paclitaxel followed by doxorubicin at disease progression versus the combination. There is no question that the response rate was higher with the combination, but at the end of the day, survival was identical in the three arms. This says to me that how you package those drugs is probably not as important as long as people are exposed to both of them in the metastatic setting.

I am philosophically more inclined toward sequential single-agent therapy in metastatic breast cancer. However, I'm fascinated by the capecitabine/ docetaxel trial. Most of the women on that trial who took docetaxel alone did not get exposure to capecitabine, and I suspect that if there had been a crossover arm, the survival would not have been much different. Having said that, I am an enthusiast about the adjuvant and neoadjuvant trials looking at the combination of capecitabine/docetaxel.

Adjuvant capecitabine trial in elderly women

I would probably be willing to put women of any age on this trial, but I think the trial focuses on elderly patients for two reasons. One is that the elderly are a research focus of Hyman Muss, the principal investigator of the trial. The other is that he thought oral therapy, which is a little less intrusive, might be more in keeping with the lifestyle issues faced by the elderly patient.

We haven’t had a lot of single-agent adjuvant therapy for quite some time, so that always gives people pause. We are revisiting whether some of our newer single agents — when given optimally — might be every bit as good as some of our combination therapies.

In the Intergroup, we are about to launch a trial in node-negative patients that is a two-by-two design involving either four or six cycles of AC versus 12 or 18 weeks of paclitaxel. The question is whether or not you can preserve the benefits of adjuvant chemotherapy with a better toxicity profile, particularly the concern about longer-term cardiotoxicity.

People were impressed by the preclinical and early clinical information, which suggested that weekly taxanes may be better than an every three-week schedule. I am interested to see whether 18 weeks of paclitaxel is the kind of therapy that you can just breeze through. It may not be quite as simple as we think.

Select publications

 

Table of Contents Top of Page

 

Home · Search

 
Editor's Note
 
Larry Norton, MD
- Select publications
 
Gabriel N Hortobagyi, MD
- Select publications
 
Nancy E Davidson, MD
- Select publications
 
 
 
Editor's office
Faculty Financial Interest or Affiliations
Home · Contact us
Terms of use and general disclaimer